1
|
Cong R, Lu C, Li X, Xu Z, Wang Y, Sun S. Tumor organoids in cancer medicine: from model systems to natural compound screening. PHARMACEUTICAL BIOLOGY 2025; 63:89-109. [PMID: 39893515 PMCID: PMC11789228 DOI: 10.1080/13880209.2025.2458149] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/26/2024] [Revised: 12/04/2024] [Accepted: 01/17/2025] [Indexed: 02/04/2025]
Abstract
CONTEXT The advent of tissue engineering and biomedical techniques has significantly advanced the development of three-dimensional (3D) cell culture systems, particularly tumor organoids. These self-assembled 3D cell clusters closely replicate the histopathological, genetic, and phenotypic characteristics of primary tissues, making them invaluable tools in cancer research and drug screening. OBJECTIVE This review addresses the challenges in developing in vitro models that accurately reflect tumor heterogeneity and explores the application of tumor organoids in cancer research, with a specific focus on the screening of natural products for antitumor therapies. METHODS This review synthesizes information from major databases, including Chemical Abstracts, Medicinal and Aromatic Plants Abstracts, ScienceDirect, Google Scholar, Scopus, PubMed and Springer Link. Publications were selected without date restrictions, using terms such as 'organoid', 'natural product', 'pharmacological', 'extract', 'nanomaterial' and 'traditional uses'. Articles related to agriculture, ecology, synthetic work or published in languages other than English were excluded. RESULTS AND CONCLUSIONS The review identifies key challenges related to the efficiency and variability of organoid generation and discusses ongoing efforts to enhance their predictive capabilities in drug screening and personalized medicine. Recent studies utilizing patient-derived organoid models for natural compound screening are highlighted, demonstrating the potential of these models in developing new classes of anticancer agents. The integration of natural products with patient-derived organoid models presents a promising approach for discovering novel anticancer compounds and elucidating their mechanisms of action.
Collapse
Affiliation(s)
- Rong Cong
- Department of General Surgery, Xiangya Hospital, Central South University, Changsha, China
| | - Can Lu
- Department of Pathology, National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
| | - Xinying Li
- Department of General Surgery, Xiangya Hospital, Central South University, Changsha, China
| | - Zhijie Xu
- Department of Pathology, National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
| | - Yaqin Wang
- Clinical Research Center for Reproduction and Genetics in Hunan Province, Reproductive and Genetic Hospital of CITIC-Xiangya, Changsha, China
| | - Shusen Sun
- College of Pharmacy and Health Sciences, Western New England University, Springfield, MA, USA
| |
Collapse
|
2
|
Chen Y, Xu H, Xiao L, Zhang M, Yan N. Single-cell RNA sequencing in the study of human retinal organoids. Exp Eye Res 2025; 256:110417. [PMID: 40320034 DOI: 10.1016/j.exer.2025.110417] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2024] [Revised: 03/26/2025] [Accepted: 05/01/2025] [Indexed: 05/10/2025]
Abstract
Single-cell RNA sequencing (scRNA-seq) has transformed the study of retinal development and diseases by enabling a detailed analysis of cellular diversity within retinal organoids (ROs). ROs generated from pluripotent stem cells mimic the essential characteristics of the human retina and provide a valuable in vitro model for investigating retinal development, cell interactions, and disease mechanisms. This review summarizes the application of scRNA-seq on RO research, emphasizing its capacity to identify distinct cell populations, uncover developmental trajectories, and reveal the molecular signatures of retinal diseases. scRNA-seq provides new insights into retinal neurogenesis, cellular diversity, and the pathophysiology of retinal degenerative diseases. This technology has enabled the identification of novel biomarkers and potential therapeutic targets. Integrating scRNA-seq with other technologies, such as spatial transcriptomics and CRISPR-based screening, can further deepen our understanding of retinal biology and improve treatment strategies.
Collapse
Affiliation(s)
- Yi Chen
- Department of Ophthalmology and Research Laboratory of Ophthalmology, West China Hospital, Sichuan University, Chengdu, Sichuan, 610041, China; Department of Ophthalmology, West China Hospital, Sichuan University, Chengdu, Sichuan, 610041, China
| | - Hanyue Xu
- Department of Ophthalmology and Research Laboratory of Ophthalmology, West China Hospital, Sichuan University, Chengdu, Sichuan, 610041, China; Department of Ophthalmology, West China Hospital, Sichuan University, Chengdu, Sichuan, 610041, China
| | - Lirong Xiao
- Department of Ophthalmology and Research Laboratory of Ophthalmology, West China Hospital, Sichuan University, Chengdu, Sichuan, 610041, China
| | - Ming Zhang
- Department of Ophthalmology, West China Hospital, Sichuan University, Chengdu, Sichuan, 610041, China.
| | - Naihong Yan
- Department of Ophthalmology and Research Laboratory of Ophthalmology, West China Hospital, Sichuan University, Chengdu, Sichuan, 610041, China.
| |
Collapse
|
3
|
Maes L, Szabó A, Van Haevermaete J, Geurs I, Dewettinck K, Vandenbroucke RE, Van Vlierberghe S, Laukens D. Digital light processing of photo-crosslinkable gelatin to create biomimetic 3D constructs serving small intestinal tissue regeneration. BIOMATERIALS ADVANCES 2025; 171:214232. [PMID: 39983500 DOI: 10.1016/j.bioadv.2025.214232] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/06/2024] [Revised: 01/27/2025] [Accepted: 02/13/2025] [Indexed: 02/23/2025]
Abstract
Regeneration of small intestinal mucosal tissue could offer a promising strategy for Crohn's disease patients suffering from chronic inflammatory damage. Here, we aimed to develop hydrogels that mirror the villi and crypts of the small intestine and exhibit a physiological stiffness of G' ~ 1.52 kPa. For this purpose, we developed gelatin-methacryloyl-aminoethyl-methacrylate (gel-MA-AEMA)-, and gelatin-methacryloyl-norbornene (gel-MA-NB)-based biomaterial inks to fabricate 3D hydrogels ("villi only" versus "crypts and villi") with digital light processing (DLP) and co-cultured Caco-2/HT29-MTX cells. Gel-MA-AEMA was selected for its higher amount of methacrylates which was hypothesized to provide superior photo-crosslinking kinetics and hence superior DLP fabrication potential while gel-MA-NB was evaluated for its selective functionalization potential with thiolated bioactive compounds following DLP processing, resulting from its incorporated NB moieties which remain unreacted during the DLP process. Both gel-MA-AEMA-, and gel-MA-NB-based hydrogels exhibited a physiologically relevant stiffness, but only the gel-MA-AEMA-based biomaterial ink could be successfully utilized for printing hydrogels encompassing villi and crypts. Paracellular permeability of small sized marker molecules in combination with transepithelial electrical resistance measurements showed the formation of a functional barrier over time on all hydrogel constructs. Transmission electron microscopy and enterocyte differentiation marker genes' expression levels revealed the superior differentiation of Caco-2 on the 3D constructs compared to 2D hydrogel sheets. In summary, while both hydrogels enhanced functional barrier formation and enterocyte differentiation, gel-MA-AEMA proved more conducive to DLP compared to gel-MA-NB. Furthermore, our study underscored the benefits of cultivating intestinal cells on soft 3D constructs, enhancing cell barrier properties and differentiation, thus providing added value over traditional 2D supports.
Collapse
Affiliation(s)
- Laure Maes
- IBD Research Unit, Department of Internal Medicine and Pediatrics, Ghent University, Ghent 9000, Belgium; Barriers in Inflammation Lab, Department of Biomedical Molecular Biology, Ghent University, Ghent 9000, Belgium; VIB-UGent Center for Inflammation Research, VIB, Ghent 9000, Belgium
| | - Anna Szabó
- Polymer Chemistry and Biomaterials Group, Centre of Macromolecular Chemistry (CMaC), Department of Organic and Macromolecular Chemistry, Ghent University, Ghent 9000, Belgium
| | - Jens Van Haevermaete
- IBD Research Unit, Department of Internal Medicine and Pediatrics, Ghent University, Ghent 9000, Belgium; Polymer Chemistry and Biomaterials Group, Centre of Macromolecular Chemistry (CMaC), Department of Organic and Macromolecular Chemistry, Ghent University, Ghent 9000, Belgium
| | - Indi Geurs
- Food Structure & Function Research Group, Department of Food Technology, Safety and Health, Ghent University, Ghent 9000, Belgium
| | - Koen Dewettinck
- Food Structure & Function Research Group, Department of Food Technology, Safety and Health, Ghent University, Ghent 9000, Belgium
| | - Roosmarijn E Vandenbroucke
- Barriers in Inflammation Lab, Department of Biomedical Molecular Biology, Ghent University, Ghent 9000, Belgium; VIB-UGent Center for Inflammation Research, VIB, Ghent 9000, Belgium
| | - Sandra Van Vlierberghe
- Polymer Chemistry and Biomaterials Group, Centre of Macromolecular Chemistry (CMaC), Department of Organic and Macromolecular Chemistry, Ghent University, Ghent 9000, Belgium.
| | - Debby Laukens
- IBD Research Unit, Department of Internal Medicine and Pediatrics, Ghent University, Ghent 9000, Belgium.
| |
Collapse
|
4
|
Li X, Zhao H, Jiang E, Liu P, Chen Y, Wang Y, Li J, Wu Y, Liu Z, Shang Z. ITGB1/FERMT1 mechanoactivation enhances CD44 characteristic stemness in oral squamous cell carcinoma via ubiquitin-dependent CK1α degradation. Oncogene 2025; 44:1530-1544. [PMID: 40044983 DOI: 10.1038/s41388-025-03317-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2024] [Revised: 01/30/2025] [Accepted: 02/18/2025] [Indexed: 05/15/2025]
Abstract
Cancer stem cells (CSCs) contribute to chemotherapy resistance and poor prognosis, posing significant challenges in the treatment of oral squamous cell carcinoma. The extracellular matrix (ECM)-constructed microenvironment remodels the niche of CSCs. Yet mechanisms by which biophysical properties of ECM relate to CSCs remain undefined. Here, our findings link ECM mechanical stimuli to CSCs phenotype transition, and propose that ECM stiffening mechanoactivates tumor cells to dedifferentiate and acquire CD44+ stem cell-like characteristics through noncanonical mechanotransduction. ITGB1 senses and transduces biomechanical signals, while FERMT1 acts as an intracellular mechanotransduction downstream, activating CSCs. Mechanistically, FERMT1 promotes the proteasomal degradation of CK1α by E3 ubiquitin ligase MIB1, thereby triggering Wnt signaling pathway. Combining targeted ECM softening with mechanotransduction inhibition strategy significantly attenuates tumor stemness and chemoresistance in vivo. Therefore, our findings highlight the role of ECM in regulating CSCs via biomechanical-dependent manner, suggesting the ECM/ITGB1/FERMT1/Wnt axis as a promising therapeutic target for CSCs therapy.
Collapse
Affiliation(s)
- Xiang Li
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan, China
| | - Hui Zhao
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan, China
- Department of Oral and Maxillofacial-Head and Neck Oncology, School of Stomatology-Hospital of Stomatology, Wuhan University, Wuhan, China
| | - Erhui Jiang
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan, China
- Department of Oral and Maxillofacial-Head and Neck Oncology, School of Stomatology-Hospital of Stomatology, Wuhan University, Wuhan, China
| | - Pan Liu
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan, China
| | - Yang Chen
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan, China
- Department of Oral and Maxillofacial Surgery, School & Hospital of Stomatology, Wuhan University, Wuhan, China
| | - Yue Wang
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan, China
| | - Ji Li
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan, China
| | - Yufei Wu
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan, China
| | - Zhenan Liu
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan, China
| | - Zhengjun Shang
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan, China.
- Department of Oral and Maxillofacial-Head and Neck Oncology, School of Stomatology-Hospital of Stomatology, Wuhan University, Wuhan, China.
| |
Collapse
|
5
|
Xie X, Chen X, Zhou J, Wang T, Yang G, Han F, Wei Z. Dynamic Hydrogels with Tunable Mechanics for 3D Organoid Derivation. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2025:e2501862. [PMID: 40434214 DOI: 10.1002/smll.202501862] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/13/2025] [Revised: 05/04/2025] [Indexed: 05/29/2025]
Abstract
The mechanical properties of the hydrogel play a pivotal role in governing the formation and development of 3D organoids in vitro. However, commonly employed natural hydrogels, such as Matrigel and other extracellular matrix (ECM)-derived products, are characterized by ill-defined and complex compositions, resulting in non-tunable mechanical properties. This limitation poses challenges in controlling organoids' developmental trajectory and 3D morphology. Although numerous synthetic hydrogels with well-defined chemical structures have recently been adopted to study organoids by modulating stiffness, advanced research emphasizes the importance of dynamic mechanical cues, such as dynamic stiffness softening and dynamic viscoelasticity, for optimal organoid derivation. These cues are essential for mimicking the dynamic physiological states of organoids during their growth. Despite their potential, the concept of dynamic hydrogels is often used interchangeably, and a systematic review is lacking to clarify this ambiguity. Furthermore, the mechanisms through which dynamic mechanical cues regulate organoid formation have not been thoroughly reported. This review endeavors to summarize and categorize dynamic hydrogels and reveal the effects of dynamic mechanics on organoid derivation. Additionally, the prospects of dynamic hydrogels in organoid derivation are deliberated to promote a more rational design of synthetic hydrogels, guiding organoid derivation and propelling organoid technology in biomedicine.
Collapse
Affiliation(s)
- Xueyong Xie
- The Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, 710049, P. R. China
- Bioinspired Engineering and Biomechanics Center (BEBC), Xi'an Jiaotong University, Xi'an, 710049, P. R. China
| | - Xuewen Chen
- The Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, 710049, P. R. China
- Bioinspired Engineering and Biomechanics Center (BEBC), Xi'an Jiaotong University, Xi'an, 710049, P. R. China
| | - Jian Zhou
- Wenzhou Institute, University of Chinese Academy of Sciences, Wenzhou, Zhejiang, 325000, P. R. China
| | - Tiansong Wang
- Department of Industrial and Systems Engineering, North Carolina State University, Raleigh, NC, 27695, USA
| | - Gen Yang
- Wenzhou Institute, University of Chinese Academy of Sciences, Wenzhou, Zhejiang, 325000, P. R. China
- School of Physics, Peking University, Beijing, 100871, P. R. China
| | - Fei Han
- The Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, 710049, P. R. China
- Bioinspired Engineering and Biomechanics Center (BEBC), Xi'an Jiaotong University, Xi'an, 710049, P. R. China
| | - Zhao Wei
- The Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, 710049, P. R. China
- Bioinspired Engineering and Biomechanics Center (BEBC), Xi'an Jiaotong University, Xi'an, 710049, P. R. China
| |
Collapse
|
6
|
Ansaryan S, Chiang YC, Liu YC, Tan J, Lorenzo-Martín LF, Lutolf MP, Tolstonog G, Altug H. Spatiotemporal Interrogation of Single Spheroids Using Multiplexed Nanoplasmonic-Fluorescence Imaging. SMALL METHODS 2025:e2500106. [PMID: 40434268 DOI: 10.1002/smtd.202500106] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/16/2025] [Revised: 04/14/2025] [Indexed: 05/29/2025]
Abstract
Advances in organoid models, as ex vivo mini-organs, and the development of screening imaging technologies have continuously driven each other forward. A complete understanding of organoids requires detailed insights into the intertwined intraorganoid and extraorganoid activities and how they change across time and space. This study introduces a multiplexed imaging platform that integrates label-free nanoplasmonic biosensing with fluorescence microscopy to offer simultaneous monitoring of dynamics occurring within and around arrays of single spheroids with spatiotemporal resolution. The label-free module employs nanoplasmonic biosensors with extraordinary optical transmission to track biomolecular secretions into the surroundings, while concurrent fluorescence imaging enables structural analysis and viability assessment. To perform multiparametric interrogation of the data from different channels over extended periods, a deep-learning-augmented image analysis is incorporated. The platform is applied to tumor spheroids to investigate vascular endothelial growth factor A secretion alongside morphometric changes and viability, showcasing its ability to capture variations in secretion and growth dynamics between untreated and drug-treated groups. This integrated approach advances comprehensive insights into organoid models and can complement existing technologies to accelerate discoveries in disease modeling and drug development.
Collapse
Affiliation(s)
- Saeid Ansaryan
- Institute of Bioengineering, École Polytechnique Fédérale de Lausanne (EPFL), Lausanne, CH-1015, Switzerland
| | - Yung-Cheng Chiang
- Institute of Bioengineering, École Polytechnique Fédérale de Lausanne (EPFL), Lausanne, CH-1015, Switzerland
| | - Yen-Cheng Liu
- Institute of Bioengineering, École Polytechnique Fédérale de Lausanne (EPFL), Lausanne, CH-1015, Switzerland
| | - Jiayi Tan
- Institute of Bioengineering, École Polytechnique Fédérale de Lausanne (EPFL), Lausanne, CH-1015, Switzerland
| | | | - Matthias P Lutolf
- Institute of Bioengineering, École Polytechnique Fédérale de Lausanne (EPFL), Lausanne, CH-1015, Switzerland
- Institute of Human Biology (IHB), Roche Pharma Research and Early Development (pRED), Roche Innovation Center Basel, Basel, CH-4070, Switzerland
| | - Genrich Tolstonog
- Department of Otolaryngology-Head and Neck Surgery, Lausanne University Hospital and University of Lausanne, Rue du Bugnon 21, Lausanne, 1011, Switzerland
- Agora Cancer Research Centre, Rue du Bugnon 25A, Lausanne, 1005, Switzerland
| | - Hatice Altug
- Institute of Bioengineering, École Polytechnique Fédérale de Lausanne (EPFL), Lausanne, CH-1015, Switzerland
| |
Collapse
|
7
|
De Luise M, Kurelac I, Coluccelli S, De Leo A, Bartoszek EM, Iorio M, Grillini M, Coadă CA, de Biase D, Marchio L, López MN, Rimmer N, Perrone AM, De Iaco P, Porcelli AM, Heinzelmann V, Martin I, Jacob F, Muraro MG, Gasparre G. Perfusion-based ex vivo culture of frozen ovarian cancer tissues with preserved tumor microenvironment. NPJ Precis Oncol 2025; 9:152. [PMID: 40410344 PMCID: PMC12102267 DOI: 10.1038/s41698-025-00941-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2024] [Accepted: 05/10/2025] [Indexed: 05/25/2025] Open
Abstract
Ovarian cancer (OC) poses significant treatment challenges due to late-stage diagnosis and a complex tumor microenvironment contributing to therapy resistance. We optimized a U-CUP perfusion-based bioreactor method to culture patient-derived primary and metastatic OC specimens, demonstrating that perfusion better preserves cancer cell viability and proliferation, both when fresh and slow-frozen tissues were used. Perfused cultures maintained key microenvironment components, including cancer-associated fibroblasts, endothelial and immune cells. Genetic analysis confirmed the retention in culture of tumor-specific driver mutations. We hence challenged ad hoc generated cisplatin-sensitive and resistant OC cells with cisplatin during growth in U-CUP, validating our system for the testing of drug response. Finally, treatment of slow-frozen OC tissues with carboplatin/paclitaxel revealed different degrees of response to treatment, as indicated by variations in tumor necrosis and number of residual PAX8+ cells, providing the bases for the prompt evaluation of OC standard chemotherapy efficacy in our ex vivo system.
Collapse
Affiliation(s)
- Monica De Luise
- Department of Medical and Surgical Sciences (DIMEC), University of Bologna, Bologna, Italy
- Centre for Applied Biomedical Research, University of Bologna, Bologna, Italy
| | - Ivana Kurelac
- Department of Medical and Surgical Sciences (DIMEC), University of Bologna, Bologna, Italy
- IRCCS Azienda Ospedaliero-Universitaria di Bologna, Bologna, Italy
| | - Sara Coluccelli
- Solid Tumor Molecular Pathology Laboratory, IRCCS Azienda Ospedaliero-Universitaria di Bologna, Bologna, Italy
| | - Antonio De Leo
- Department of Medical and Surgical Sciences (DIMEC), University of Bologna, Bologna, Italy
- Solid Tumor Molecular Pathology Laboratory, IRCCS Azienda Ospedaliero-Universitaria di Bologna, Bologna, Italy
| | - Ewelina M Bartoszek
- Microscopy Core Facility, Department of Biomedicine, University of Basel, Basel, Switzerland
| | - Maria Iorio
- Department of Medical and Surgical Sciences (DIMEC), University of Bologna, Bologna, Italy
- Tissue Engineering, Department of Biomedicine, University of Basel and University Hospital of Basel, Basel, Switzerland
- Ovarian Cancer Research, Department of Biomedicine, University of Basel and University Hospital of Basel, Basel, Switzerland
| | - Marco Grillini
- Pathology Unit, IRCCS Azienda Ospedaliero-Universitaria di Bologna, Bologna, Italy
| | | | - Dario de Biase
- Solid Tumor Molecular Pathology Laboratory, IRCCS Azienda Ospedaliero-Universitaria di Bologna, Bologna, Italy
- Department of Pharmacy and Biotechnology (FABIT), University of Bologna, Bologna, Italy
| | - Lorena Marchio
- Department of Medical and Surgical Sciences (DIMEC), University of Bologna, Bologna, Italy
| | - Mónica Núñez López
- Ovarian Cancer Research, Department of Biomedicine, University of Basel and University Hospital of Basel, Basel, Switzerland
| | - Natalie Rimmer
- Ovarian Cancer Research, Department of Biomedicine, University of Basel and University Hospital of Basel, Basel, Switzerland
| | - Anna Myriam Perrone
- Department of Medical and Surgical Sciences (DIMEC), University of Bologna, Bologna, Italy
- Division of Gynecologic Oncology, IRCCS Azienda Ospedaliero-Universitaria di Bologna, Bologna, Italy
| | - Pierandrea De Iaco
- Department of Medical and Surgical Sciences (DIMEC), University of Bologna, Bologna, Italy
- Division of Gynecologic Oncology, IRCCS Azienda Ospedaliero-Universitaria di Bologna, Bologna, Italy
| | - Anna Maria Porcelli
- IRCCS Azienda Ospedaliero-Universitaria di Bologna, Bologna, Italy
- Department of Pharmacy and Biotechnology (FABIT) and Centre for Applied Biomedical Research (CRBA), University of Bologna, Bologna, Italy
| | - Viola Heinzelmann
- Ovarian Cancer Research, Department of Biomedicine, University of Basel and University Hospital of Basel, Basel, Switzerland
| | - Ivan Martin
- Tissue Engineering, Department of Biomedicine, University of Basel and University Hospital of Basel, Basel, Switzerland
- Department of Biomedical Engineering, University of Basel, Basel, Switzerland
| | - Francis Jacob
- Ovarian Cancer Research, Department of Biomedicine, University of Basel and University Hospital of Basel, Basel, Switzerland
| | - Manuele Giuseppe Muraro
- Tissue Engineering, Department of Biomedicine, University of Basel and University Hospital of Basel, Basel, Switzerland.
| | - Giuseppe Gasparre
- Department of Medical and Surgical Sciences (DIMEC), University of Bologna, Bologna, Italy.
- Centre for Applied Biomedical Research, University of Bologna, Bologna, Italy.
- Centro Studi E Ricerca Sulle Neoplasie Ginecologiche (CSR), University of Bologna, Bologna, Italy.
| |
Collapse
|
8
|
Tonini L, Ahn C. Latest Advanced Techniques for Improving Intestinal Organoids Limitations. Stem Cell Rev Rep 2025:10.1007/s12015-025-10894-9. [PMID: 40388043 DOI: 10.1007/s12015-025-10894-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/06/2025] [Indexed: 05/20/2025]
Abstract
Intestinal organoids are valuable tools across different disciplines, from a clinical aspect to the biomedical research, providing a unique perspective on the complexity of the gastrointestinal system. They are alternatives to common cell lines as they can offer insights into architectural functionality and reduce the use of animal models. A deeper understanding of their organoid characteristics is required to harness their full potential. Despite their beneficial uses and multiple advantages, organoids have limitations that remain unaddressed. This review aims to elucidate the principal limitations of intestinal organoids, investigate structural defects such as the deficiency in a vascularized and lymphatic system, and absence of the microbiome, restrictions in mimicking the physiological gut model, including the lack of an acid-neutralizing system or a shortage of digestive enzymes, and the difficulties in their long-term maintenance and polarity accessibility. Development of innovative techniques to address these limitations will lead to improve in vivo recapitulation and pioneering further advancements in this field.
Collapse
Affiliation(s)
- Lisa Tonini
- Laboratory of Veterinary Physiology, College of Veterinary Medicine, Jeju National University, Jeju, 63243, Republic of Korea
| | - Changhwan Ahn
- Laboratory of Veterinary Physiology, College of Veterinary Medicine, Jeju National University, Jeju, 63243, Republic of Korea.
- Veterinary Medical Research Institute, Jeju National University, Jeju, 63243, Republic of Korea.
| |
Collapse
|
9
|
Kumar D, Gupta S, Gupta V, Tanwar R, Chandel A. Engineering the Future of Regenerative Medicines in Gut Health with Stem Cell-Derived Intestinal Organoids. Stem Cell Rev Rep 2025:10.1007/s12015-025-10893-w. [PMID: 40380985 DOI: 10.1007/s12015-025-10893-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/05/2025] [Indexed: 05/19/2025]
Abstract
The advent of intestinal organoids, three-dimensional structures derived from stem cells, has significantly advanced the field of biology by providing robust in vitro models that closely mimic the architecture and functionality of the human intestine. These organoids, generated from induced pluripotent stem cells (iPSCs), embryonic stem cells (ESCs), or adult stem cells, possess remarkable capabilities for self-renewal, differentiation into diverse intestinal cell types, and functional recapitulation of physiological processes, including nutrient absorption, epithelial barrier integrity, and host-microbe interactions. The utility of intestinal organoids has been extensively demonstrated in disease modeling, drug screening, and personalized medicine. Notable examples include iPSC-derived organoids, which have been effectively employed to model enteric infections, and ESC-derived organoids, which have provided critical insights into fetal intestinal development. Patient-derived organoids have emerged as powerful tools for investigating personalized therapeutics and regenerative interventions for conditions such as inflammatory bowel disease (IBD), cystic fibrosis, and colorectal cancer. Preclinical studies involving transplantation of human intestinal organoids into murine models have shown promising outcomes, including functional integration, epithelial restoration, and immune system interactions. Despite these advancements, several challenges persist, particularly in achieving reproducibility, scalability, and maturation of organoids, which hinder their widespread clinical translation. Addressing these limitations requires the establishment of standardized protocols for organoid generation, culture, storage, and analysis to ensure reproducibility and comparability of findings across studies. Nevertheless, intestinal organoids hold immense promise for transforming our understanding of gastrointestinal pathophysiology, enhancing drug development pipelines, and advancing personalized medicine. By bridging the gap between preclinical research and clinical applications, these organoids represent a paradigm shift in the exploration of novel therapeutic strategies and the investigation of gut-associated diseases.
Collapse
Affiliation(s)
- Dinesh Kumar
- School of Pharmacy, Desh Bhagat University, Mandi Gobindgarh, Punjab, India.
| | - Sonia Gupta
- Swami Devi Dyal Group of Professional Institute, Panchkula, India
| | - Vrinda Gupta
- School of Pharmacy, Desh Bhagat University, Mandi Gobindgarh, Punjab, India
| | - Rajni Tanwar
- School of Pharmacy, Desh Bhagat University, Mandi Gobindgarh, Punjab, India
| | - Anchal Chandel
- School of Pharmacy, Desh Bhagat University, Mandi Gobindgarh, Punjab, India
| |
Collapse
|
10
|
Song Y, Seitz M, Kowalczewski A, Mai NY, Jain E, Yang H, Ma Z. Mechanically and Chemically Defined PEG Hydrogels Improve Reproducibility in Human Cardioid Development. Adv Healthc Mater 2025:e2403997. [PMID: 40376871 DOI: 10.1002/adhm.202403997] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2024] [Revised: 03/22/2025] [Indexed: 05/18/2025]
Abstract
Cardioids are 3D self-organized heart organoids directly derived from induced pluripotent stem cells (hiPSCs) aggregates. The growth and culture of cardioids is either conducted in suspension culture or heavily relies on Matrigel encapsulation. Despite the significant advancements in cardioid technology, reproducibility remains a major challenge, limiting their widespread use in both basic research and translational applications. Here, for the first time, we employed synthetic, matrix metalloproteinase (MMP)-degradable polyethylene glycol (PEG)-based hydrogels to define the effect of mechanical and biochemical cues on cardioid development. Successful cardiac differentiation is demonstrated in all the hydrogel conditions, while cardioid cultured in optimized PEG hydrogel (3 wt.% PEG-2mM RGD) underwent similar morphological development and comparable tissue functions to those cultured in Matrigel. Matrix stiffness and cell adhesion motif play a critical role in cardioid development, nascent chamber formation, contractile physiology, and endothelial cell gene enrichment. More importantly, synthetic hydrogel improved the reproducibility in cardioid properties compared to traditional suspension culture and Matrigel encapsulation. Therefore, PEG-based hydrogel has the potential to be used as an alternative to Matrigel for human cardioid culture in a variety of clinical applications including cell therapy and tissue engineering.
Collapse
Affiliation(s)
- Yuanhui Song
- Department of Biomedical & Chemical Engineering, Syracuse University, Syracuse, NY, 13244, USA
- BioInspired Institute for Material and Living Systems, Syracuse University, Syracuse, NY, 13244, USA
| | - Michael Seitz
- Department of Biomedical & Chemical Engineering, Syracuse University, Syracuse, NY, 13244, USA
- BioInspired Institute for Material and Living Systems, Syracuse University, Syracuse, NY, 13244, USA
| | - Andrew Kowalczewski
- Department of Biomedical & Chemical Engineering, Syracuse University, Syracuse, NY, 13244, USA
- BioInspired Institute for Material and Living Systems, Syracuse University, Syracuse, NY, 13244, USA
| | - Nhu Y Mai
- Department of Biomedical & Chemical Engineering, Syracuse University, Syracuse, NY, 13244, USA
- BioInspired Institute for Material and Living Systems, Syracuse University, Syracuse, NY, 13244, USA
| | - Era Jain
- Department of Biomedical & Chemical Engineering, Syracuse University, Syracuse, NY, 13244, USA
- BioInspired Institute for Material and Living Systems, Syracuse University, Syracuse, NY, 13244, USA
| | - Huaxiao Yang
- Department of Biomedical Engineering, University of North Texas, Denton, TX, 76205, USA
| | - Zhen Ma
- Department of Biomedical & Chemical Engineering, Syracuse University, Syracuse, NY, 13244, USA
- BioInspired Institute for Material and Living Systems, Syracuse University, Syracuse, NY, 13244, USA
| |
Collapse
|
11
|
Liu Y, Gilchrist AE, Johansson PK, Guan Y, Deras JD, Liu YC, Ceva S, Huang MS, Navarro RS, Enejder A, Peltz G, Heilshorn SC. Engineered Hydrogels for Organoid Models of Human Nonalcoholic Fatty Liver Disease. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025:e17332. [PMID: 40364726 DOI: 10.1002/advs.202417332] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/20/2024] [Revised: 04/22/2025] [Indexed: 05/15/2025]
Abstract
Nonalcoholic fatty liver disease (NAFLD) is characterized by increased lipid accumulation and excessive deposition of extracellular matrix (ECM) that results in tissue stiffening. The potential interplay between matrix stiffness and hepatocyte lipid accumulation during NAFLD has not been established. Here, an in vitro NAFLD model is developed using chemically defined, engineered hydrogels and human induced pluripotent stem cell-derived hepatic organoids (HOs). Specifically, dynamic covalent chemistry crosslinking, along with transient small molecule competitors, are used to create dynamic stiffening hydrogels that enable the reproducible culture of HOs. Within matrices that mimic the stiffness of healthy to diseased tissue (≈1-6 kPa), lipid droplet accumulation in HOs is triggered by exposure to an NAFLD-associated free fatty acid. These NAFLD model suggests that higher stiffness microenvironments result in increased hepatic lipid droplet accumulation, increased expression of fibrosis markers, and increased metabolic dysregulation. By targeting the ROCK mechanosignaling pathway, the synergy between matrix stiffness and lipid droplet accumulation is disrupted. The in vitro model of NAFLD has the potential to understand the role of mechanosignaling in disease progression and identify new pathways for therapeutic intervention.
Collapse
Affiliation(s)
- Yueming Liu
- Department of Materials Science and Engineering, Stanford University, Stanford, CA, 94305, USA
| | - Aidan E Gilchrist
- Department of Biomedical Engineering, University of California, Davis, CA, 95616, USA
| | - Patrik K Johansson
- Department of Materials Science and Engineering, Stanford University, Stanford, CA, 94305, USA
| | - Yuan Guan
- Department of Anesthesiology, Pain and Perioperative Medicine, Stanford University School of Medicine, Stanford, CA, 94305, USA
| | - Jaydon D Deras
- Department of Chemical Engineering, Stanford University, Stanford, CA, 94305, USA
| | - Yu-Chung Liu
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI, 48109, USA
| | - Sofia Ceva
- Department of Biology, Stanford University, Stanford, CA, 94305, USA
| | - Michelle S Huang
- Department of Chemical Engineering, Stanford University, Stanford, CA, 94305, USA
| | - Renato S Navarro
- Department of Materials Science and Engineering, Stanford University, Stanford, CA, 94305, USA
| | - Annika Enejder
- Department of Materials Science and Engineering, Stanford University, Stanford, CA, 94305, USA
| | - Gary Peltz
- Department of Anesthesiology, Pain and Perioperative Medicine, Stanford University School of Medicine, Stanford, CA, 94305, USA
| | - Sarah C Heilshorn
- Department of Materials Science and Engineering, Stanford University, Stanford, CA, 94305, USA
| |
Collapse
|
12
|
Jabri A, Taftafa B, Mhannayeh A, Alsharif M, Abbad T, Ahmed S, Alshehri EA, Elsalti A, Khan J, Mir TA, Yaqinuddin A. Cardiac Tissue Engineering for Translational Cardiology: From In Vitro Models to Regenerative Therapies. Bioengineering (Basel) 2025; 12:518. [PMID: 40428138 PMCID: PMC12109445 DOI: 10.3390/bioengineering12050518] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2025] [Revised: 05/07/2025] [Accepted: 05/09/2025] [Indexed: 05/29/2025] Open
Abstract
Cardiovascular diseases (CVD) are the primary cause of death and disability around the world. Over the past decades, several conventional model systems based on two-dimensional (3D) monolayer cultures or experimental animals have been adopted to dissect and understand heart diseases in order to develop treatment modalities. However, traditional models exhibit several limitations in recapitulating human-specific key physiological and pathological characteristics, which highlights the necessity of developing physiologically relevant models. In recent years, tissue engineering approaches have been extensively employed to generate revolutionary three-dimensional (3D) cardiac models. In particular, the combined use of various bioengineering strategies and cellular reprogramming approaches has facilitated the development of various models. This review presents an overview of different approaches (bioprinting, scaffolding, and electrospinning) for creating bioengineered cardiac tissue models. Next, a broad survey of recent research related to the modeling of various cardiac diseases is presented. Finally, current challenges and future directions are proposed to foster further developments in the field of cardiac tissue engineering.
Collapse
Affiliation(s)
- Abdullah Jabri
- College of Medicine, Alfaisal University, Riyadh 11533, Saudi Arabia; (A.J.); (B.T.); (A.M.); (M.A.); (T.A.); (J.K.)
| | - Bader Taftafa
- College of Medicine, Alfaisal University, Riyadh 11533, Saudi Arabia; (A.J.); (B.T.); (A.M.); (M.A.); (T.A.); (J.K.)
| | - Abdulaziz Mhannayeh
- College of Medicine, Alfaisal University, Riyadh 11533, Saudi Arabia; (A.J.); (B.T.); (A.M.); (M.A.); (T.A.); (J.K.)
| | - Mohamed Alsharif
- College of Medicine, Alfaisal University, Riyadh 11533, Saudi Arabia; (A.J.); (B.T.); (A.M.); (M.A.); (T.A.); (J.K.)
| | - Tasnim Abbad
- College of Medicine, Alfaisal University, Riyadh 11533, Saudi Arabia; (A.J.); (B.T.); (A.M.); (M.A.); (T.A.); (J.K.)
| | - Sana Ahmed
- Tissue/Organ Bioengineering & BioMEMS Laboratory, Organ Transplant Centre of Excellence (TR&I-Dpt), King Faisal Specialist Hospital and Research Centre, Riyadh 11211, Saudi Arabia; (S.A.); (E.A.A.)
| | - Eman A. Alshehri
- Tissue/Organ Bioengineering & BioMEMS Laboratory, Organ Transplant Centre of Excellence (TR&I-Dpt), King Faisal Specialist Hospital and Research Centre, Riyadh 11211, Saudi Arabia; (S.A.); (E.A.A.)
| | - Abdulrahman Elsalti
- International School of Medicine, Istanbul Medipol University, Istanbul 34810, Turkey;
| | - Jibran Khan
- College of Medicine, Alfaisal University, Riyadh 11533, Saudi Arabia; (A.J.); (B.T.); (A.M.); (M.A.); (T.A.); (J.K.)
| | - Tanveer Ahmad Mir
- College of Medicine, Alfaisal University, Riyadh 11533, Saudi Arabia; (A.J.); (B.T.); (A.M.); (M.A.); (T.A.); (J.K.)
- Tissue/Organ Bioengineering & BioMEMS Laboratory, Organ Transplant Centre of Excellence (TR&I-Dpt), King Faisal Specialist Hospital and Research Centre, Riyadh 11211, Saudi Arabia; (S.A.); (E.A.A.)
| | - Ahmed Yaqinuddin
- College of Medicine, Alfaisal University, Riyadh 11533, Saudi Arabia; (A.J.); (B.T.); (A.M.); (M.A.); (T.A.); (J.K.)
| |
Collapse
|
13
|
Kopyeva I, Bretherton RC, Ayers JL, Yu M, Grady WM, DeForest CA. Matrix Stiffness and Biochemistry Govern Colorectal Cancer Cell Growth and Signaling in User-Programmable Synthetic Hydrogels. ACS Biomater Sci Eng 2025; 11:2810-2823. [PMID: 40304602 DOI: 10.1021/acsbiomaterials.4c01632] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/02/2025]
Abstract
Colorectal cancer (CRC) studies in vitro have been conducted almost exclusively on 2D cell monolayers or suspension spheroid cultures. Though these platforms have shed light on many important aspects of CRC biology, they fail to recapitulate essential cell-matrix interactions that often define in vivo function. Toward filling this knowledge gap, synthetic hydrogel biomaterials with user-programmable matrix mechanics and biochemistry have gained popularity for culturing cells in a more physiologically relevant 3D context. Here, using a poly(ethylene glycol)-based hydrogel model, we systematically assess the role of matrix stiffness and fibronectin-derived RGDS adhesive peptide presentation on CRC colony morphology and proliferation. Highlighting platform generalizability, we demonstrate that these hydrogels can support the viability and promote spontaneous spheroid or multicellular aggregate formation of six CRC cell lines that are commonly utilized in biomedical research. These gels are engineered to be fully degradable via a "biologically invisible" sortase-mediated reaction, enabling the triggered recovery of single cells and spheroids for downstream analysis. Using these platforms, we establish that substrate mechanics play a significant role in colony growth: soft conditions (∼300 Pa) encourage robust colony formation, whereas stiffer (∼2 kPa) gels severely restrict growth. Tuning the RGDS concentration did not affect the colony morphology. Additionally, we observe that epidermal growth factor receptor (EGFR) signaling in Caco-2 cells is influenced by adhesion ligand identity─whether the adhesion peptide was derived from collagen type I (DGEA) or fibronectin (RGDS)─with DGEA yielding a marked decrease in the level of downstream protein kinase phosphorylation. Taken together, this study introduces a versatile method to culture and probe CRC cell-matrix interactions within engineered 3D biomaterials.
Collapse
Affiliation(s)
- Irina Kopyeva
- Department of Bioengineering, University of Washington, Seattle 98105, Washington, United States
- Institute of Stem Cell & Regenerative Medicine, University of Washington, Seattle 98105, Washington, United States
| | - Ross C Bretherton
- Department of Bioengineering, University of Washington, Seattle 98105, Washington, United States
- Institute of Stem Cell & Regenerative Medicine, University of Washington, Seattle 98105, Washington, United States
| | - Jessica L Ayers
- Translational Science and Therapeutics Division, Fred Hutchinson Cancer Center, Seattle 98109, Washington, United States
| | - Ming Yu
- Translational Science and Therapeutics Division, Fred Hutchinson Cancer Center, Seattle 98109, Washington, United States
| | - William M Grady
- Translational Science and Therapeutics Division, Fred Hutchinson Cancer Center, Seattle 98109, Washington, United States
- Department of Internal Medicine, University of Washington, Seattle 98105, Washington, United States
| | - Cole A DeForest
- Department of Bioengineering, University of Washington, Seattle 98105, Washington, United States
- Institute of Stem Cell & Regenerative Medicine, University of Washington, Seattle 98105, Washington, United States
- Department of Chemical Engineering, University of Washington, Seattle 98105, Washington, United States
- Molecular Engineering & Sciences Institute, University of Washington, Seattle 98105, Washington, United States
- Department of Chemistry, University of Washington, Seattle 98105, Washington, United States
- Institute for Protein Design, University of Washington, Seattle 98105, Washington, United States
| |
Collapse
|
14
|
Teleanu MV, Schneider A, Ball CR, Leber MF, Stange C, Krieghoff-Henning E, Beck K, Heilig CE, Kreutzfeldt S, Kuster B, Lipka DB, Fröhling S. Celebrating Ulrik Ringborg: Multi-Omics-Based Patient Stratification for Precision Cancer Treatment. Biomolecules 2025; 15:693. [PMID: 40427586 PMCID: PMC12108785 DOI: 10.3390/biom15050693] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2025] [Revised: 05/03/2025] [Accepted: 05/08/2025] [Indexed: 05/29/2025] Open
Abstract
Precision oncology is becoming a mainstay in the standard of care for cancer patients. Recent technological advancements have significantly lowered the cost of various tumor profiling approaches, broadening the reach of molecular diagnostics and improving patient access to precision oncology. In parallel, drug development and discovery pipelines continue to evolve, driving targeted therapeutic options forward. Yet, not all patients harboring actionable molecular alterations respond to these interventions, and existing therapies do not cover the entire spectrum of potential molecular targets. In this review, we examine the current suite of omics technologies employed in clinical settings and underscore their roles in deepening our understanding of tumor biology and optimizing patient stratification for targeted treatments. We also highlight relevant precision oncology trials and share our own experiences using multi-omics data within a molecular tumor board framework. Finally, we discuss areas for future exploration aimed at propelling precision oncology to new heights.
Collapse
Affiliation(s)
- Maria-Veronica Teleanu
- Division of Translational Medical Oncology, German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany
- National Center for Tumor Diseases (NCT), NCT Heidelberg, a Partnership Between DKFZ and Heidelberg University Hospital, 69120 Heidelberg, Germany
- German Cancer Consortium (DKTK), Core Center Heidelberg, 69120 Heidelberg, Germany
| | - Annika Schneider
- School of Life Sciences, Technical University Munich, 85354 Freising, Germany
- German Cancer Consortium (DKTK), Partner Site Munich, 80336 Munich, Germany
| | - Claudia R. Ball
- Department of Translational Medical Oncology, National Center for Tumor Diseases (NCT)/University Cancer Center Dresden, a Partnership Between DKFZ, Faculty of Medicine and University Hospital Carl Gustav Carus, Dresden University of Technology (TUD), and Helmholtz-Zentrum Dresden-Rossendorf, 01307 Dresden, Germany
- Translational Medical Oncology, Faculty of Medicine and University Hospital Carl Gustav Carus, Dresden University of Technology (TUD), 01069 Dresden, Germany
- Faculty of Biology, Dresden University of Technology (TUD), 01217 Dresden, Germany
- German Cancer Consortium (DKTK), Partner Site Dresden, 01307 Dresden, Germany
| | - Mathias Felix Leber
- National Center for Tumor Diseases (NCT), NCT Heidelberg, a Partnership Between DKFZ and Heidelberg University Hospital, 69120 Heidelberg, Germany
- Department of Medical Oncology, Heidelberg University Hospital, 69120 Heidelberg, Germany
- Clinical Cooperation Unit Virotherapy, German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany
| | - Christoph Stange
- School of Life Sciences, Technical University Munich, 85354 Freising, Germany
- German Cancer Consortium (DKTK), Partner Site Munich, 80336 Munich, Germany
| | - Eva Krieghoff-Henning
- Division of Translational Medical Oncology, German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany
- National Center for Tumor Diseases (NCT), NCT Heidelberg, a Partnership Between DKFZ and Heidelberg University Hospital, 69120 Heidelberg, Germany
- Division of Personalized Medical Oncology, German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany
- Department of Personalized Oncology, DKFZ-Hector Cancer Institute, University Medical Center Mannheim, Medical Faculty Mannheim, Heidelberg University, 68167 Mannheim, Germany
- Department of Personalized Oncology, University Medical Center Mannheim, Medical Faculty Mannheim, Heidelberg University, 68167 Mannheim, Germany
| | - Katja Beck
- Division of Translational Medical Oncology, German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany
- National Center for Tumor Diseases (NCT), NCT Heidelberg, a Partnership Between DKFZ and Heidelberg University Hospital, 69120 Heidelberg, Germany
- German Cancer Consortium (DKTK), Core Center Heidelberg, 69120 Heidelberg, Germany
| | - Christoph E. Heilig
- Division of Translational Medical Oncology, German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany
- National Center for Tumor Diseases (NCT), NCT Heidelberg, a Partnership Between DKFZ and Heidelberg University Hospital, 69120 Heidelberg, Germany
- German Cancer Consortium (DKTK), Core Center Heidelberg, 69120 Heidelberg, Germany
| | - Simon Kreutzfeldt
- Division of Translational Medical Oncology, German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany
- National Center for Tumor Diseases (NCT), NCT Heidelberg, a Partnership Between DKFZ and Heidelberg University Hospital, 69120 Heidelberg, Germany
- German Cancer Consortium (DKTK), Core Center Heidelberg, 69120 Heidelberg, Germany
| | - Bernhard Kuster
- School of Life Sciences, Technical University Munich, 85354 Freising, Germany
- German Cancer Consortium (DKTK), Partner Site Munich, 80336 Munich, Germany
| | - Daniel B. Lipka
- Division of Translational Medical Oncology, German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany
- National Center for Tumor Diseases (NCT), NCT Heidelberg, a Partnership Between DKFZ and Heidelberg University Hospital, 69120 Heidelberg, Germany
- German Cancer Consortium (DKTK), Core Center Heidelberg, 69120 Heidelberg, Germany
| | - Stefan Fröhling
- Division of Translational Medical Oncology, German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany
- National Center for Tumor Diseases (NCT), NCT Heidelberg, a Partnership Between DKFZ and Heidelberg University Hospital, 69120 Heidelberg, Germany
- German Cancer Consortium (DKTK), Core Center Heidelberg, 69120 Heidelberg, Germany
- Institute of Human Genetics, Heidelberg University, 69120 Heidelberg, Germany
| |
Collapse
|
15
|
Li K, He Y, Jin X, Jin K, Qian J. Reproducible extracellular matrices for tumor organoid culture: challenges and opportunities. J Transl Med 2025; 23:497. [PMID: 40312683 PMCID: PMC12044958 DOI: 10.1186/s12967-025-06349-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2025] [Accepted: 03/03/2025] [Indexed: 05/03/2025] Open
Abstract
Tumor organoid models have emerged as valuable 3D in vitro systems to study cancer behavior in a physiologically relevant environment. The composition and architecture of the extracellular matrix (ECM) play critical roles in tumor organoid culture by influencing the tumor microenvironment and tumor behavior. Traditional matrices such as Matrigel and collagen, have been widely used, but their batch-to-batch variability and limited tunability hinder their reproducibility and broader applications. To address these challenges, researchers have turned to synthetic/engineered matrices and biopolymer-based matrices, which offer precise tunability, reproducibility, and chemically defined compositions. However, these matrices also present challenges of their own. In this review, we explore the significance of ECMs in tumor organoid culture, discuss the limitations of commonly used matrices, and highlight recent advancements in engineered/synthetic matrices for improved tumor organoid modeling.
Collapse
Affiliation(s)
- Kan Li
- School of Basic Medical Sciences, Health Science Center, Ningbo University, Ningbo, 315211, China
| | - Yibo He
- Department of Surgical Oncology, Hangzhou Cancer Hospital, Hangzhou, Zhejiang, 310006, China
- Department of Breast Surgery, Affiliated Hangzhou First People'S Hospital, School of Medicine, Westlake University, Hangzhou, Zhejiang, 310006, China
| | - Xue Jin
- Center for Clinical Pharmacy, Cancer Center, Department of Pharmacy, Zhejiang Provincial People'S Hospital (Affiliated People'S Hospital, Hangzhou Medical College), Hangzhou, Zhejiang, 310014, China
| | - Ketao Jin
- Department of Colorectal and Anal Surgery, The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Chinese Medicine), Hangzhou, Zhejiang, 310003, China.
| | - Jun Qian
- Department of Colorectal Surgery, Xinchang People'S Hospital, Affiliated Xinchang Hosptial, Wenzhou Medical University, Xinchang, Zhejiang, 312500, China.
| |
Collapse
|
16
|
Li S, Ren J, Wu J, Xia Z, Li Y, Li C, Cao W. Establishment and molecular characterisation of patient-derived organoids for primary central nervous system lymphoma. Leukemia 2025; 39:1169-1183. [PMID: 40102628 DOI: 10.1038/s41375-025-02562-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2024] [Revised: 02/20/2025] [Accepted: 03/05/2025] [Indexed: 03/20/2025]
Abstract
Primary central nervous system lymphoma (PCNSL) exhibits substantial intratumoural and intertumoural heterogeneity, complicating the development of effective treatment methods. Existing in vitro models fail to simulate the cellular and mutational diversity of native tumours and require prolonged generation times. Therefore, we developed a culture method for patient-derived PCNSL organoids (CLOs) and evaluated the organoids through extensive molecular characterisation, histopathological analysis, single-nucleus RNA sequencing, bulk RNA sequencing and whole-exome sequencing. These CLOs accurately mimicked the histological attributes, gene expression landscapes and mutational profiles of their original tumours. Single-nucleus RNA sequencing also revealed that CLOs maintained cell-type heterogeneity and the molecular signatures of their original tumours. CLOs were generated within 2 weeks, demonstrating rapid development and reliability. Therapeutic profiling was performed on three selected CLOs treated with four standard drugs. The CLOs exhibited specific sensitivity to methotrexate, and resistance to dexamethasone, ibrutinib and rituximab, suggesting that CLOs may be valuable tools for reflecting drug sensitivities. Taken together, these results emphasise that CLOs effectively emulate the key characteristics of PCNSL, increasing the understanding of the genetic landscape of this complex disease. CLOs provide a rapid and reliable platform for exploring individualised treatment strategies, potentially accelerating the transition of research findings to clinical practice.
Collapse
Affiliation(s)
- Shengjie Li
- Department of Clinical Laboratory, Eye & ENT Hospital, Shanghai Medical College, Fudan University, Shanghai, China.
- Department of Neurosurgery, Huashan Hospital, Shanghai Medical College, Fudan University, Shanghai, China.
- National Center for Neurological Disorders, Shanghai, China.
- Shanghai Key Laboratory of Brain Function Restoration and Neural Regeneration, Shanghai, China.
- Neurosurgical Institute of Fudan University, Shanghai, China.
- Shanghai Clinical Medical Center of Neurosurgery, Shanghai, China.
| | - Jun Ren
- Department of Clinical Laboratory, Eye & ENT Hospital, Shanghai Medical College, Fudan University, Shanghai, China
| | - Jianing Wu
- Department of Clinical Laboratory, Eye & ENT Hospital, Shanghai Medical College, Fudan University, Shanghai, China
| | - Zuguang Xia
- Department of Medical Oncology, Fudan University Shanghai Cancer Center, Fudan University, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Yingzhu Li
- Department of Clinical Laboratory, Eye & ENT Hospital, Shanghai Medical College, Fudan University, Shanghai, China
| | - Chengxun Li
- Department of Otolaryngology, Eye & ENT Hospital, Fudan University, Shanghai, China.
| | - Wenjun Cao
- Department of Clinical Laboratory, Eye & ENT Hospital, Shanghai Medical College, Fudan University, Shanghai, China.
| |
Collapse
|
17
|
Han J, Jeong H, Choi J, Kim H, Kwon T, Myung K, Park K, Park JI, Sánchez S, Jung D, Yu CS, Song IH, Shim J, Myung S, Kang H, Park T. Bioprinted Patient-Derived Organoid Arrays Capture Intrinsic and Extrinsic Tumor Features for Advanced Personalized Medicine. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025; 12:e2407871. [PMID: 40151904 PMCID: PMC12120747 DOI: 10.1002/advs.202407871] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/11/2024] [Revised: 02/11/2025] [Indexed: 03/29/2025]
Abstract
Heterogeneity and the absence of a tumor microenvironment (TME) in traditional patient-derived organoid (PDO) cultures limit their effectiveness for clinical use. Here, Embedded Bioprinting-enabled Arrayed PDOs (Eba-PDOs) featuring uniformly arrayed colorectal cancer (CRC) PDOs within a recreated TME is presented. This model faithfully reproduces critical TME attributes, including elevated matrix stiffness (≈7.5 kPa) and hypoxic conditions found in CRC. Transcriptomic and immunofluorescence microscopy analysis reveal that Eba-PDOs more accurately represent actual tissues compared to traditional PDOs. Furthermore, Eba-PDO effectively capture the variability of CEACAM5 expression-a critical CRC marker-across different patients, correlating with patient classification and differential responses to 5-fluorouracil treatment. This method achieves an uniform size and shape within PDOs from the same patient while preserving distinct morphological features among those from different individuals. These features of Eba-PDO enable the efficient development of a label-free, morphology-based predictive model using supervised learning, enhancing its suitability for clinical applications. Thus, this approach to PDO bioprinting is a promising tool for generating personalized tumor models and advancing precision medicine.
Collapse
Affiliation(s)
- Jonghyeuk Han
- Department of Biomedical EngineeringUlsan National Institute of Science and Technology (UNIST)Ulsan44919Republic of Korea
- Wallace H. Coulter Department of Biomedical EngineeringEmory University School of Medicine & Georgia Institute of TechnologyAtlantaGA30332USA
| | - Hye‐Jin Jeong
- Department of Biomedical EngineeringUlsan National Institute of Science and Technology (UNIST)Ulsan44919Republic of Korea
- Center for Genome EngineeringInstitute for Basic ScienceDaejeon34126Republic of Korea
| | - Jeonghan Choi
- Department of Biomedical EngineeringUlsan National Institute of Science and Technology (UNIST)Ulsan44919Republic of Korea
| | - Hyeonseo Kim
- Department of Biomedical EngineeringUlsan National Institute of Science and Technology (UNIST)Ulsan44919Republic of Korea
| | - Taejoon Kwon
- Department of Biomedical EngineeringUlsan National Institute of Science and Technology (UNIST)Ulsan44919Republic of Korea
- Center for Genomic IntegrityInstitute for Basic ScienceUlsan44919Republic of Korea
| | - Kyungjae Myung
- Department of Biomedical EngineeringUlsan National Institute of Science and Technology (UNIST)Ulsan44919Republic of Korea
- Center for Genomic IntegrityInstitute for Basic ScienceUlsan44919Republic of Korea
| | - Kyemyung Park
- Graduate School of Health Science and Technology and Department of Biomedical EngineeringUlsan National Institute of Science and TechnologyUlsan44919Republic of Korea
| | - Jung In Park
- Department of Biomedical EngineeringUlsan National Institute of Science and Technology (UNIST)Ulsan44919Republic of Korea
| | - Samuel Sánchez
- Institute for Bioengineering of Catalonia (IBEC)The Barcelona Institute for Science and Technology (BIST)Barcelona08028Spain
- Catalan Institute for Research and Advanced Studies (ICREA)Barcelona08010Spain
| | - Deok‐Beom Jung
- Digestive Diseases Research CenterUniversity of Ulsan College of MedicineSeoul05505Republic of Korea
| | - Chang Sik Yu
- Division of Colon and Rectal SurgeryDepartment of SurgeryAsan Medical CenterUniversity of Ulsan College of MedicineSeoul05505Republic of Korea
| | - In Ho Song
- Division of Colon and Rectal SurgeryDepartment of SurgeryAsan Medical CenterUniversity of Ulsan College of MedicineSeoul05505Republic of Korea
| | - Jin‐Hyung Shim
- Research InstituteT&R Biofab Co. Ltd.Siheung15111Republic of Korea
- Department of Mechanical EngineeringTech University of KoreaSiheung15073Republic of Korea
| | - Seung‐Jae Myung
- Digestive Diseases Research CenterUniversity of Ulsan College of MedicineSeoul05505Republic of Korea
- Department of GastroenterologyAsan Medical CenterUniversity of Ulsan College of MedicineSeoul05505Republic of Korea
- EDIS BiotechSeoul05505Republic of Korea
| | - Hyun‐Wook Kang
- Department of Biomedical EngineeringUlsan National Institute of Science and Technology (UNIST)Ulsan44919Republic of Korea
| | - Tae‐Eun Park
- Department of Biomedical EngineeringUlsan National Institute of Science and Technology (UNIST)Ulsan44919Republic of Korea
| |
Collapse
|
18
|
Yavitt FM, Khang A, Bera K, McNally DL, Blatchley MR, Gallagher AP, Klein OD, Castillo-Azofeifa D, Dempsey PJ, Anseth KS. Engineered epithelial curvature controls Paneth cell localization in intestinal organoids. CELL BIOMATERIALS 2025; 1:100046. [PMID: 40270579 PMCID: PMC12013698 DOI: 10.1016/j.celbio.2025.100046] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/25/2025]
Abstract
The cellular organization within organoid models is important to regulate tissue specific function, yet few engineering approaches can control or direct cellular organization. Here, a photodegradable hydrogel is used to create softened regions that direct crypt formation within intestinal organoids, where the dimensions of the photosoftened regions generate predictable and defined crypt architectures. Guided by in vivo metrics of crypt morphology, this photopatterning method is used to control the width and length of in vitro organoid crypts, which ultimately defines the curvature of the epithelium. By tracking expression of differentiated Paneth cell markers in real-time, we show that epithelial curvature directs the localization of Paneth cells within engineered crypts, providing user-directed control over organoid functionality. We anticipate that our improved control over organoid architecture and thus Paneth cell localization will lead to more consistent in vitro organoid models for both mechanistic studies and translational applications.
Collapse
Affiliation(s)
- F. Max Yavitt
- Department of Chemical and Biological Engineering, University of Colorado Boulder, Boulder, CO, 80303, USA
- BioFrontiers Institute, University of Colorado Boulder, Boulder, CO, 80303, USA
| | - Alex Khang
- Department of Chemical and Biological Engineering, University of Colorado Boulder, Boulder, CO, 80303, USA
- BioFrontiers Institute, University of Colorado Boulder, Boulder, CO, 80303, USA
| | - Kaustav Bera
- Department of Chemical and Biological Engineering, University of Colorado Boulder, Boulder, CO, 80303, USA
- BioFrontiers Institute, University of Colorado Boulder, Boulder, CO, 80303, USA
| | - Delaney L. McNally
- Department of Chemical and Biological Engineering, University of Colorado Boulder, Boulder, CO, 80303, USA
| | - Michael R. Blatchley
- Department of Chemical and Biological Engineering, University of Colorado Boulder, Boulder, CO, 80303, USA
- BioFrontiers Institute, University of Colorado Boulder, Boulder, CO, 80303, USA
| | - Aaron P. Gallagher
- Department of Orofacial Sciences and Program in Craniofacial Biology, University of California, San Francisco, San Francisco, CA, 90089, USA
- School of Medicine, University of California, San Francisco, San Francisco, CA, 94143, USA
| | - Ophir D. Klein
- Department of Orofacial Sciences and Program in Craniofacial Biology, University of California, San Francisco, San Francisco, CA, 90089, USA
- Department of Pediatrics and Guerin Children’s, Cedars-Sinai Medical Center, Los Angeles, CA, 90505, USA
- Department of Pediatrics and Institute for Human Genetics, University of California, San Francisco, San Francisco, CA, 94143, USA
| | - David Castillo-Azofeifa
- Department of Regenerative Medicine, Genentech, Inc., South San Francisco, California, 94080, USA
| | - Peter J. Dempsey
- Section of Developmental Biology, Department of Pediatrics, University of Colorado, Denver, CO, 80045, USA
| | - Kristi S. Anseth
- Department of Chemical and Biological Engineering, University of Colorado Boulder, Boulder, CO, 80303, USA
- BioFrontiers Institute, University of Colorado Boulder, Boulder, CO, 80303, USA
| |
Collapse
|
19
|
Clerkin S, Singh K, Davis JL, Treacy NJ, Krupa I, Reynaud EG, Lees RM, Needham SR, MacWhite-Begg D, Wychowaniec JK, Brougham DF, Crean J. Tuneable gelatin methacryloyl (GelMA) hydrogels for the directed specification of renal cell types for hiPSC-derived kidney organoid maturation. Biomaterials 2025; 322:123349. [PMID: 40315627 DOI: 10.1016/j.biomaterials.2025.123349] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2024] [Revised: 02/14/2025] [Accepted: 04/15/2025] [Indexed: 05/04/2025]
Abstract
Diabetic Kidney Disease (DKD) represents a significant global health burden and is recognised as the leading cause of end-stage renal disease. Kidney organoids derived from human induced Pluripotent Stem Cells (hiPSCs) have the potential to transform how we model renal disease and may provide personalised replacement tissues for patients with renal failure. However, kidney organoids remain poorly reproducible, and are structurally and functionally immature. Three-dimensional cultures that more appropriately mimic the complexity of the in vivo microenvironment are required to improve organoid maturation and structural authenticity. Here, we describe the application of semi-synthetic Gelatin Methacryloyl (GelMA) hydrogels as extracellular support matrices for the differentiation of hiPSC-derived kidney organoids. Hydrogels of defined mechanical strengths were generated by varying the concentration of GelMA solution in the presence of low concentration photo-initiator. After confirming a high level of mechanical stability of the hydrogels over extended culture periods, their effect on kidney organoid maturation was investigated. Organoids differentiated within GelMA hydrogels generated typical renal cell types including podocytes, tubular epithelia, renal interstitial cells, and some nascent vascularisation. Interestingly, kidney organoids derived within hydrogels that closely approximate the stiffness of the adult human kidney (∼5000-10,000 Pa) demonstrated improved podocyte maturation and were shown to upregulate renal vesicle-associated genes at an earlier stage following encapsulation when compared to organoids derived within softer hydrogels (∼400 Pa). A model of TGFβ-induced injury was also developed to investigate the influence of the mechanical environment in propagating early, fibrotic-like features of DKD within organoids. Growth within the softer matrix was shown to reduce pSMAD3 expression following TGFβ1 treatment, and accordingly ameliorate the expression of the myofibroblast marker α-Smooth Muscle Actin (α-SMA). This work demonstrates the suitability of GelMA hydrogels as mechanically-stable, highly-tuneable, batch-to-batch reproducible three-dimensional supports for hiPSC-derived kidney organoid growth and differentiation, and further substantiates the role of the biophysical environment in guiding processes of cell fate determination and organoid maturation.
Collapse
Affiliation(s)
- Shane Clerkin
- UCD School of Biomolecular and Biomedical Science, UCD Conway Institute of Biomolecular and Biomedical Research, University College Dublin, Belfield, Dublin 4, Ireland
| | - Krutika Singh
- UCD School of Chemistry, University College Dublin, Belfield, Dublin 4, Ireland
| | - Jessica L Davis
- UCD School of Biomolecular and Biomedical Science, UCD Conway Institute of Biomolecular and Biomedical Research, University College Dublin, Belfield, Dublin 4, Ireland
| | - Niall J Treacy
- UCD School of Biomolecular and Biomedical Science, UCD Conway Institute of Biomolecular and Biomedical Research, University College Dublin, Belfield, Dublin 4, Ireland
| | - Ivan Krupa
- UCD School of Biomolecular and Biomedical Science, UCD Conway Institute of Biomolecular and Biomedical Research, University College Dublin, Belfield, Dublin 4, Ireland
| | - Emmanuel G Reynaud
- UCD School of Biomolecular and Biomedical Science, UCD Conway Institute of Biomolecular and Biomedical Research, University College Dublin, Belfield, Dublin 4, Ireland
| | - Robert M Lees
- Science and Technology Research Council Central Laser Facility (STFC-CLF), Rutherford Appleton Laboratory, Harwell, Didcot, OX11 0DE, United Kingdom
| | - Sarah R Needham
- Science and Technology Research Council Central Laser Facility (STFC-CLF), Rutherford Appleton Laboratory, Harwell, Didcot, OX11 0DE, United Kingdom
| | - Delphi MacWhite-Begg
- UCD School of Biomolecular and Biomedical Science, UCD Conway Institute of Biomolecular and Biomedical Research, University College Dublin, Belfield, Dublin 4, Ireland
| | - Jacek K Wychowaniec
- UCD School of Chemistry, University College Dublin, Belfield, Dublin 4, Ireland
| | - Dermot F Brougham
- UCD School of Chemistry, University College Dublin, Belfield, Dublin 4, Ireland
| | - John Crean
- UCD School of Biomolecular and Biomedical Science, UCD Conway Institute of Biomolecular and Biomedical Research, University College Dublin, Belfield, Dublin 4, Ireland.
| |
Collapse
|
20
|
Su X, Wang M, Yuan R, Guo L, Han Y, Huang C, Li A, Kaplan DL, Wang X. Organoids in Dynamic Culture: Microfluidics and 3D Printing Technologies. ACS Biomater Sci Eng 2025. [PMID: 40248908 DOI: 10.1021/acsbiomaterials.4c02245] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/19/2025]
Abstract
With the rapid advancement of biomaterials and tissue engineering technologies, organoid research and its applications have made significant strides. Organoids are increasingly utilized in pharmacology, regenerative medicine, and precision clinical medicine. Current trends in organoid research are moving toward multifunctional composite three-dimensional cultivation and dynamic cultivation strategies. Key technologies driving this evolution, including 3D printing and microfluidics, continue to impact new areas of discovery and clinical relevance. This review provides a systematic overview of these emerging trends, discussing the strengths and limitations of these critical technologies and offering insight and research directions for professionals working in the organoid field.
Collapse
Affiliation(s)
- Xin Su
- College of Basic Medical Sciences, Dalian Medical University, Dalian, China 116044
| | - Mingqi Wang
- College of Basic Medical Sciences, Dalian Medical University, Dalian, China 116044
| | - Ruqiang Yuan
- College of Basic Medical Sciences, Dalian Medical University, Dalian, China 116044
- Advanced Institute for Medical Sciences, Dalian Medical University, Dalian, China 116044
| | - Lina Guo
- College of Basic Medical Sciences, Dalian Medical University, Dalian, China 116044
| | - Yinhe Han
- College of Basic Medical Sciences, Dalian Medical University, Dalian, China 116044
| | - Chun Huang
- College of Basic Medical Sciences, Dalian Medical University, Dalian, China 116044
| | - Ang Li
- College of Basic Medical Sciences, Dalian Medical University, Dalian, China 116044
| | - David L Kaplan
- Department of Biomedical Engineering, Tufts University, Medford, Massachusetts 02155, United States
| | - Xiuli Wang
- College of Basic Medical Sciences, Dalian Medical University, Dalian, China 116044
| |
Collapse
|
21
|
Kuhn MR, Wolcott EA, Langer EM. Developments in gastrointestinal organoid cultures to recapitulate tissue environments. Front Bioeng Biotechnol 2025; 13:1521044. [PMID: 40313639 PMCID: PMC12043594 DOI: 10.3389/fbioe.2025.1521044] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2024] [Accepted: 03/21/2025] [Indexed: 05/03/2025] Open
Abstract
Culture platforms that closely mimic the spatial architecture, cellular diversity, and extracellular matrix composition of native tissues can serve as invaluable tools for a range of scientific discovery and biomedical applications. Organoids have emerged as a promising alternative to both traditional 2D cell culture and animal models, offering a physiologically relevant 3D culture system for studying human cell biology. Organoids provide a manipulable platform to investigate organ development and function as well as to model patient-specific phenotypes. This mini review examines various methods used for culturing organoids to model normal and disease conditions in gastrointestinal tissues. We focus on how the matrix composition and media formulations can impact cell signaling, altering the baseline cellular phenotypes as well as response to perturbations. We discuss future directions for optimizing organoid culture conditions to improve basic and translational potential.
Collapse
Affiliation(s)
- Madeline R. Kuhn
- Cancer Early Detection Advanced Research Center, Knight Cancer Institute, Oregon Health and Science University, Portland, OR, United States
- Division of Oncological Sciences, Oregon Health and Science University, Portland, OR, United States
| | - Emma A. Wolcott
- Cancer Early Detection Advanced Research Center, Knight Cancer Institute, Oregon Health and Science University, Portland, OR, United States
- Division of Oncological Sciences, Oregon Health and Science University, Portland, OR, United States
| | - Ellen M. Langer
- Cancer Early Detection Advanced Research Center, Knight Cancer Institute, Oregon Health and Science University, Portland, OR, United States
- Division of Oncological Sciences, Oregon Health and Science University, Portland, OR, United States
- Brenden-Colson Center for Pancreatic Care, Oregon Health and Science University, Portland, OR, United States
| |
Collapse
|
22
|
Wang Y, Sun X, Lu B, Zhang D, Yin Y, Liu S, Chen L, Zhang Z. Current applications, future Perspectives and challenges of Organoid technology in oral cancer research. Eur J Pharmacol 2025; 993:177368. [PMID: 39947346 DOI: 10.1016/j.ejphar.2025.177368] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2024] [Revised: 01/31/2025] [Accepted: 02/10/2025] [Indexed: 02/20/2025]
Abstract
Oral cancer poses significant health risks with an increasing incidence annually. Despite advancements in treatment methods, their efficacy is frequently constrained by cancer heterogeneity and drug resistance, leading to minimal improvement in the 5-year survival rate. Therefore, there is a critical need for new treatment methods leaded by representative preclinical research models. Compared to other models, organoids can more precisely simulate the tissue structure, genetic characteristics, and tumor microenvironment (TME) of in vivo tumors, exhibiting high tumor specificity. This makes organoid technology a valuable tool in investigating tumor development, mechanisms of metastasis, drug screening, prediction of clinical responses, and personalized patient treatment. Moreover, integrating organoid technology with other biotechnologies could expand its applications in tissue regeneration. Although organoid technology is increasingly utilized in oral cancer research, a systematic review in this field is absent. This paper is to bridge the gap by reviewing the development and current status of organoid research, highlighting its applications, future prospects, and challenges in oral cancer. It aims to provide novel insights into the role of organoids in precision treatment and regenerative medicine for oral cancer.
Collapse
Affiliation(s)
- Yunyi Wang
- Department of Oral and Maxillofacial Surgery, Stomatological Hospital, School of Stomatology, Southern Medical University, Guangzhou, China
| | - Xiang Sun
- Department of Oral and Maxillofacial Surgery, Stomatological Hospital, School of Stomatology, Southern Medical University, Guangzhou, China
| | - Bingxu Lu
- Department of Oral and Maxillofacial Surgery, Stomatological Hospital, School of Stomatology, Southern Medical University, Guangzhou, China
| | - Danya Zhang
- Department of Oral and Maxillofacial Surgery, Stomatological Hospital, School of Stomatology, Southern Medical University, Guangzhou, China
| | - Yaping Yin
- Department of Oral and Maxillofacial Surgery, Stomatological Hospital, School of Stomatology, Southern Medical University, Guangzhou, China
| | - Shuguang Liu
- Department of Oral and Maxillofacial Surgery, Stomatological Hospital, School of Stomatology, Southern Medical University, Guangzhou, China.
| | - Lei Chen
- Department of Burn, Wound Repair & Reconstruction, The First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China; Guangdong Provincial Engineering Technology Research Center of Burn and Wound Accurate Diagnosis and Treatment Key Technology and Series of Products, Sun Yat-Sen University, Guangzhou, China; Institute of Precision Medicine, The First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, China.
| | - Zhaoqiang Zhang
- Department of Oral and Maxillofacial Surgery, Stomatological Hospital, School of Stomatology, Southern Medical University, Guangzhou, China.
| |
Collapse
|
23
|
Xie D, Chen B, Wang W, Guo W, Sun Z, Wang L, Shi B, Song Y, Su M. Nanocomposite Hydrogels and Micro/Nanostructures for Printing Organoids. ACS NANO 2025; 19:12458-12466. [PMID: 40162603 DOI: 10.1021/acsnano.4c17587] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/02/2025]
Abstract
Organoids are 3D artificial miniature organs composed of a cluster of self-renewing and self-organizing cells in vitro, which mimic the functions of real organs. Nanotechnologies, including the preparation of nanomaterials and the fabrication of micro/nanostructures, have been proven to promote cell proliferation, guide cell differentiation, and regulate cell self-organization, showing great promise in engineering organoids. In this Perspective, different types of nanocomposite hydrogels for organoid culture are summarized, the effects of micro/nanostructures on organoid growth and development are discussed, and 3D bioprinting technologies for constructing organoid models are introduced.
Collapse
Affiliation(s)
- Daixi Xie
- Key Laboratory of Green Printing, Institute of Chemistry, Chinese Academy of Sciences, Beijing Engineering Research Center of Nanomaterials for Green Printing Technology, Beijing National Laboratory for Molecular Sciences (BNLMS), Zhongguancun North First Street 2, 100190 Beijing, P. R. China
- University of Chinese Academy of Sciences, Yuquan Road No. 19A, 100049 Beijing, P. R. China
| | - Bingda Chen
- Key Laboratory of Green Printing, Institute of Chemistry, Chinese Academy of Sciences, Beijing Engineering Research Center of Nanomaterials for Green Printing Technology, Beijing National Laboratory for Molecular Sciences (BNLMS), Zhongguancun North First Street 2, 100190 Beijing, P. R. China
| | - Wenqing Wang
- Key Laboratory of Green Printing, Institute of Chemistry, Chinese Academy of Sciences, Beijing Engineering Research Center of Nanomaterials for Green Printing Technology, Beijing National Laboratory for Molecular Sciences (BNLMS), Zhongguancun North First Street 2, 100190 Beijing, P. R. China
- University of Chinese Academy of Sciences, Yuquan Road No. 19A, 100049 Beijing, P. R. China
| | - Wenjing Guo
- University of Chinese Academy of Sciences, Yuquan Road No. 19A, 100049 Beijing, P. R. China
| | - Zhiyuan Sun
- Macao Institute of Materials Science and Engineering, Faculty of Innovation Engineering, Macau University of Science and Technology, Taipa, Macao 999078, China
| | - Long Wang
- Department of Orthopedics, the Fourth Medical Center of Chinese PLA General Hospital, Beijing 100048, China
| | - Bin Shi
- Department of Organ Transplantation, the Third Medical Center of PLA General Hospital, Beijing 100039, China
| | - Yanlin Song
- Key Laboratory of Green Printing, Institute of Chemistry, Chinese Academy of Sciences, Beijing Engineering Research Center of Nanomaterials for Green Printing Technology, Beijing National Laboratory for Molecular Sciences (BNLMS), Zhongguancun North First Street 2, 100190 Beijing, P. R. China
- University of Chinese Academy of Sciences, Yuquan Road No. 19A, 100049 Beijing, P. R. China
| | - Meng Su
- Key Laboratory of Green Printing, Institute of Chemistry, Chinese Academy of Sciences, Beijing Engineering Research Center of Nanomaterials for Green Printing Technology, Beijing National Laboratory for Molecular Sciences (BNLMS), Zhongguancun North First Street 2, 100190 Beijing, P. R. China
- University of Chinese Academy of Sciences, Yuquan Road No. 19A, 100049 Beijing, P. R. China
| |
Collapse
|
24
|
Xu H, Kang J, Gao X, Lan Y, Li M. Towards a Better Understanding of the Human Health Risk of Per- and Polyfluoroalkyl Substances Using Organoid Models. Bioengineering (Basel) 2025; 12:393. [PMID: 40281753 PMCID: PMC12025065 DOI: 10.3390/bioengineering12040393] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2025] [Revised: 03/16/2025] [Accepted: 03/28/2025] [Indexed: 04/29/2025] Open
Abstract
The ubiquitous presence of per- and polyfluoroalkyl substances (PFAS) in the environment has garnered global public concern. Epidemiological studies have proved that exposure to PFAS is associated with human health risks. Although evidence demonstrated the toxic mechanisms of PFAS based on animal models and traditional cell cultures, their limitations in inter-species differences and lack of human-relevant microenvironments hinder the understanding of health risks from PFAS exposure. There is an increasing necessity to explore alternative methodologies that can effectively evaluate human health risks. Human organoids derived from stem cells accurately mimic the sophisticated and multicellular structures of native human organs, providing promising models for toxicology research. Advanced organoids combined with innovative technologies are expected to improve understanding of the breadth and depth of PFAS toxicity.
Collapse
Affiliation(s)
- Haoan Xu
- School of Life Sciences and Technology, Tongji University, Shanghai 200120, China;
| | - Jiahui Kang
- Key Lab of Visual Damage and Regeneration & Restoration of Chongqing, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing 400038, China;
| | - Xue Gao
- Key Laboratory of Biorheological Science and Technology, Ministry of Education, College of Bioengineering, Chongqing University, Chongqing 400030, China;
| | - Yingying Lan
- Institute of Burn Research, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing 400038, China
| | - Minghui Li
- Key Lab of Visual Damage and Regeneration & Restoration of Chongqing, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing 400038, China;
- Key Laboratory of Biorheological Science and Technology, Ministry of Education, College of Bioengineering, Chongqing University, Chongqing 400030, China;
| |
Collapse
|
25
|
Liu K, Chen X, Fan Z, Ren F, Liu J, Hu B. From organoids to organoids-on-a-chip: Current applications and challenges in biomedical research. Chin Med J (Engl) 2025; 138:792-807. [PMID: 39994843 PMCID: PMC11970821 DOI: 10.1097/cm9.0000000000003535] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2024] [Indexed: 02/26/2025] Open
Abstract
ABSTRACT The high failure rates in clinical drug development based on animal models highlight the urgent need for more representative human models in biomedical research. In response to this demand, organoids and organ chips were integrated for greater physiological relevance and dynamic, controlled experimental conditions. This innovative platform-the organoids-on-a-chip technology-shows great promise in disease modeling, drug discovery, and personalized medicine, attracting interest from researchers, clinicians, regulatory authorities, and industry stakeholders. This review traces the evolution from organoids to organoids-on-a-chip, driven by the necessity for advanced biological models. We summarize the applications of organoids-on-a-chip in simulating physiological and pathological phenotypes and therapeutic evaluation of this technology. This section highlights how integrating technologies from organ chips, such as microfluidic systems, mechanical stimulation, and sensor integration, optimizes organoid cell types, spatial structure, and physiological functions, thereby expanding their biomedical applications. We conclude by addressing the current challenges in the development of organoids-on-a-chip and offering insights into the prospects. The advancement of organoids-on-a-chip is poised to enhance fidelity, standardization, and scalability. Furthermore, the integration of cutting-edge technologies and interdisciplinary collaborations will be crucial for the progression of organoids-on-a-chip technology.
Collapse
Affiliation(s)
- Kailun Liu
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China
- Key Laboratory of Organ Regeneration and Reconstruction, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China
| | - Xiaowei Chen
- Institute of Biophysics, Chinese Academy of Sciences, Beijing 100101, China
| | - Zhen Fan
- Institute of Biophysics, Chinese Academy of Sciences, Beijing 100101, China
| | - Fei Ren
- State Key Lab of Processors, Institute of Computing Technology, Chinese Academy of Sciences, Beijing 100190, China
| | - Jing Liu
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China
- Key Laboratory of Organ Regeneration and Reconstruction, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China
| | - Baoyang Hu
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China
- Key Laboratory of Organ Regeneration and Reconstruction, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China
- Beijing Institute for Stem Cell and Regenerative Medicine, Beijing 100101 China
| |
Collapse
|
26
|
Courbot O, Elosegui-Artola A. The role of extracellular matrix viscoelasticity in development and disease. NPJ BIOLOGICAL PHYSICS AND MECHANICS 2025; 2:10. [PMID: 40191103 PMCID: PMC11968406 DOI: 10.1038/s44341-025-00014-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 08/30/2024] [Accepted: 02/14/2025] [Indexed: 04/09/2025]
Abstract
For several decades, research has studied the influence of the extracellular matrix (ECM) mechanical properties in cell response, primarily emphasising its elasticity as the main determinant of cell and tissue behaviour. However, the ECM is not purely elastic; it is viscoelastic. ECM viscoelasticity has now emerged as a major regulator of collective cell dynamics. This review highlights recent findings on the role of ECM viscoelasticity in development and pathology.
Collapse
Affiliation(s)
- Olivia Courbot
- Cell and Tissue Mechanobiology Laboratory, The Francis Crick Institute, London, UK
- Department of Physics, King’s College London, London, UK
| | - Alberto Elosegui-Artola
- Cell and Tissue Mechanobiology Laboratory, The Francis Crick Institute, London, UK
- Department of Physics, King’s College London, London, UK
| |
Collapse
|
27
|
Magne L, Bugarin F, Ferrand A. How to Study the Mechanobiology of Intestinal Epithelial Organoids? A Review of Culture Supports, Imaging Techniques, and Analysis Methods. Biol Cell 2025; 117:e70003. [PMID: 40223609 PMCID: PMC11995250 DOI: 10.1111/boc.70003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2025] [Revised: 02/28/2025] [Accepted: 03/06/2025] [Indexed: 04/15/2025]
Abstract
Mechanobiology studies how mechanical forces influence biological processes at different scales, both in homeostasis and in pathology. Organoids, 3D structures derived from stem cells, are particularly relevant tools for modeling tissues and organs in vitro. They currently constitute one of the most suitable models for mechanobiology studies. This review provides an overview of existing or applicable approaches to organoids for mechanical studies. We first present the different types of culture supports, including hydrogels and organ-on-chip. We then discuss advanced imaging techniques, particularly suitable for studying the physical properties of cells, allowing the visualization of mechanical forces and cellular responses. We also describe the approaches and tools available to observe the organoids by microscopy. Finally, we present analytical methods, including computational models and biophysical measurement approaches, which facilitate the quantification of mechanical interactions. This review aims to provide the most comprehensive overview possible of the methods, instrumentations, and tools available to conduct a mechanobiological study on organoids.
Collapse
Affiliation(s)
- Léa Magne
- Institut de Recherche en Santé DigestiveUniversité de Toulouse, INSERM, INRAE, ENVT, UPSToulouseFrance
- Institut Clément AderUniversité Fédérale de Toulouse Midi‐Pyrénées, CNRS, UPS, INSA, ISAE‐SUPAEROToulouseFrance
| | - Florian Bugarin
- Institut Clément AderUniversité Fédérale de Toulouse Midi‐Pyrénées, CNRS, UPS, INSA, ISAE‐SUPAEROToulouseFrance
| | - Audrey Ferrand
- Institut de Recherche en Santé DigestiveUniversité de Toulouse, INSERM, INRAE, ENVT, UPSToulouseFrance
| |
Collapse
|
28
|
Xu X, Zhang Y, Huang G, Perekatt A, Wang Y, Chen L. Advances and applications of gut organoids: modeling intestinal diseases and therapeutic development. LIFE MEDICINE 2025; 4:lnaf012. [PMID: 40276096 PMCID: PMC12018802 DOI: 10.1093/lifemedi/lnaf012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 09/21/2024] [Accepted: 03/04/2025] [Indexed: 04/26/2025]
Abstract
Gut organoids are 3D cellular structures derived from adult or pluripotent stem cells, capable of closely replicating the physiological properties of the gut. These organoids serve as powerful tools for studying gut development and modeling the pathogenesis of intestinal diseases. This review provides an in-depth exploration of technological advancements and applications of gut organoids, with a focus on their construction methods. Additionally, the potential applications of gut organoids in disease modeling, microenvironmental simulation, and personalized medicine are summarized. This review aims to offer perspectives and directions for understanding the mechanisms of intestinal health and disease as well as for developing innovative therapeutic strategies.
Collapse
Affiliation(s)
- Xiaoting Xu
- School of Life Science and Technology, Key Laboratory of Developmental Genes and Human Disease, Southeast University, Nanjing 210031, China
| | - Yuping Zhang
- School of Life Science and Technology, Key Laboratory of Developmental Genes and Human Disease, Southeast University, Nanjing 210031, China
| | - Guoxin Huang
- Clinical Research Center, Shantou Key Laboratory of Basic and Translational Research of Malignant Tumor, Shantou Central Hospital, Shantou 515041, China
| | - Ansu Perekatt
- Department of Chemistry and Chemical Biology, Stevens Institute of Technology, Hoboken, NJ 07030, United States
| | - Yan Wang
- Center for Translation Medicine Research and Development, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China
| | - Lei Chen
- School of Life Science and Technology, Key Laboratory of Developmental Genes and Human Disease, Southeast University, Nanjing 210031, China
- Institute of Microphysiological Systems, Southeast University, Nanjing 211189, China
| |
Collapse
|
29
|
Seth P, Friedrichs J, Limasale YDP, Fertala N, Freudenberg U, Zhang Y, Lampel A, Werner C. Interpenetrating Polymer Network Hydrogels with Tunable Viscoelasticity and Proteolytic Cleavability to Direct Stem Cells In Vitro. Adv Healthc Mater 2025; 14:e2402656. [PMID: 39506429 PMCID: PMC11973941 DOI: 10.1002/adhm.202402656] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2024] [Revised: 10/18/2024] [Indexed: 11/08/2024]
Abstract
The dynamic nature of cellular microenvironments, regulated by the viscoelasticity and enzymatic cleavage of the extracellular matrix, remains challenging to emulate in engineered synthetic biomaterials. To address this, a novel platform of cell-instructive hydrogels is introduced, composed of two concurrently forming interpenetrating polymer networks (IPNs). These IPNs consist of the same basic building blocks - four-armed poly(ethylene glycol) and the sulfated glycosaminoglycan (sGAG) heparin - are cross-linked through either chemical or physical interactions, allowing for precise and selective tuning of the hydrogel's stiffness, viscoelasticity, and proteolytic cleavability. The studies of the individual and combined effects of these parameters on stem cell behavior revealed that human mesenchymal stem cells exhibited increased spreading and Yes-associated protein transcriptional activity in more viscoelastic and cleavable sGAG-IPN hydrogels. Furthermore, human induced pluripotent stem cell (iPSC) cysts displayed enhanced lumen formation, growth, and pluripotency maintenance when cultured in sGAG-IPN hydrogels with higher viscoelasticity. Inhibition studies emphasized the pivotal roles of actin dynamics and matrix metalloproteinase activity in iPSC cyst morphology, which varied with the viscoelastic properties of the hydrogels. Thus, the introduced sGAG-IPN hydrogel platform offers a powerful methodology for exogenous stem cell fate control.
Collapse
Affiliation(s)
- Prannoy Seth
- Leibniz Institute of Polymer Research DresdenMax Bergmann Center of Biomaterials Dresden01069DresdenGermany
| | - Jens Friedrichs
- Leibniz Institute of Polymer Research DresdenMax Bergmann Center of Biomaterials Dresden01069DresdenGermany
| | - Yanuar Dwi Putra Limasale
- Leibniz Institute of Polymer Research DresdenMax Bergmann Center of Biomaterials Dresden01069DresdenGermany
| | - Nicole Fertala
- Leibniz Institute of Polymer Research DresdenMax Bergmann Center of Biomaterials Dresden01069DresdenGermany
| | - Uwe Freudenberg
- Leibniz Institute of Polymer Research DresdenMax Bergmann Center of Biomaterials Dresden01069DresdenGermany
| | - Yixin Zhang
- Cluster of Excellence Physics of Lifeand B CUBE – Center for Molecular BioengineeringDresden University of Technology01307DresdenGermany
| | - Ayala Lampel
- Shmunis School of Biomedicine and Cancer ResearchGeorge S. Wise Faculty of Life SciencesCenter for Nanoscience and Nanotechnology Sagol Center for Regenerative Biotechnologyand Center for the Physics and Chemistry of Living Systems Tel Aviv UniversityTel Aviv69978Israel
| | - Carsten Werner
- Leibniz Institute of Polymer Research DresdenMax Bergmann Center of Biomaterials Dresden01069DresdenGermany
- Center for Regenerative Therapies Dresdenand Cluster of Excellence Physics of LifeDresden University of Technology01062DresdenGermany
| |
Collapse
|
30
|
Yang Q, Li M, Xiao Z, Feng Y, Lei L, Li S. A New Perspective on Precision Medicine: The Power of Digital Organoids. Biomater Res 2025; 29:0171. [PMID: 40129676 PMCID: PMC11931648 DOI: 10.34133/bmr.0171] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2024] [Revised: 02/21/2025] [Accepted: 03/04/2025] [Indexed: 03/26/2025] Open
Abstract
Precision medicine is a personalized medical model based on the individual's genome, phenotype, and lifestyle that provides tailored treatment plans for patients. In this context, tumor organoids, a 3-dimensional preclinical model based on patient-derived tumor cell self-organization, combined with digital analysis methods, such as high-throughput sequencing and image processing technology, can be used to analyze the genome, transcriptome, and cellular heterogeneity of tumors, so as to accurately track and assess the growth process, genetic characteristics, and drug responsiveness of tumor organoids, thereby facilitating the implementation of precision medicine. This interdisciplinary approach is expected to promote the innovation of cancer diagnosis and enhance personalized treatment. In this review, the characteristics and culture methods of tumor organoids are summarized, and the application of multi-omics, such as bioinformatics and artificial intelligence, and the digital methods of organoids in precision medicine research are discussed. Finally, this review explores the main causes and potential solutions for the bottleneck in the clinical translation of digital tumor organoids, proposes the prospects of multidisciplinary cooperation and clinical transformation to narrow the gap between laboratory and clinical settings, and provides references for research and development in this field.
Collapse
Affiliation(s)
- Qian Yang
- Department of Otorhinolaryngology Head and Neck Surgery, The Second Xiangya Hospital,
Central South University, Changsha 410011, Hunan, China
| | - Mengmeng Li
- Department of Otorhinolaryngology Head and Neck Surgery, The Second Xiangya Hospital,
Central South University, Changsha 410011, Hunan, China
| | - Zian Xiao
- Department of Otorhinolaryngology Head and Neck Surgery, The Second Xiangya Hospital,
Central South University, Changsha 410011, Hunan, China
| | - Yekai Feng
- Department of Otorhinolaryngology Head and Neck Surgery, The Second Xiangya Hospital,
Central South University, Changsha 410011, Hunan, China
| | - Lanjie Lei
- Key Laboratory of Artificial Organs and Computational Medicine in Zhejiang Province, Institute of Translational Medicine,
Zhejiang Shuren University, Hangzhou 310015, Zhejiang, China
| | - Shisheng Li
- Department of Otorhinolaryngology Head and Neck Surgery, The Second Xiangya Hospital,
Central South University, Changsha 410011, Hunan, China
| |
Collapse
|
31
|
Wang K, Wang Y, Han J, Liang Z, Zhang W, Li X, Chen J, Wang L. Biofabrication and simulation techniques for gut-on-a-chip. Biofabrication 2025; 17:022011. [PMID: 39965538 DOI: 10.1088/1758-5090/adb7c1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2024] [Accepted: 02/18/2025] [Indexed: 02/20/2025]
Abstract
Biomimetic gut models show promise for enhancing our understanding of intestinal disorder pathogenesis and accelerating therapeutic strategy development. Currentin vitromodels predominantly comprise traditional static cell culture and animal models. Static cell culture lacks the precise control of the complex microenvironment governing human intestinal function. Animal models provide greater microenvironment complexity but fail to accurately replicate human physiological conditions due to interspecies differences. As the available models do not accurately reflect the microphysiological environment and functions of the human intestine, their applications are limited. An optimal approach to intestinal modeling is yet to be developed, but the field will probably benefit from advances in biofabrication techniques. This review highlights biofabrication strategies for constructing biomimetic intestinal models and research approaches for simulating key intestinal physiological features. We also discuss potential biomedical applications of these models and provide an outlook on multi-scale intestinal modeling.
Collapse
Affiliation(s)
- Ke Wang
- School of Mechanical Engineering, Qilu University of Technology (Shandong Academy of Sciences), Jinan 250353, People's Republic of China
- Shandong Institute of Mechanical Design and Research, Jinan 250353, People's Republic of China
| | - Yushen Wang
- School of Mechanical Engineering, Qilu University of Technology (Shandong Academy of Sciences), Jinan 250353, People's Republic of China
- Shandong Institute of Mechanical Design and Research, Jinan 250353, People's Republic of China
| | - Junlei Han
- School of Mechanical Engineering, Qilu University of Technology (Shandong Academy of Sciences), Jinan 250353, People's Republic of China
- Shandong Institute of Mechanical Design and Research, Jinan 250353, People's Republic of China
| | - Zhixiang Liang
- School of Mechanical Engineering, Qilu University of Technology (Shandong Academy of Sciences), Jinan 250353, People's Republic of China
- Shandong Institute of Mechanical Design and Research, Jinan 250353, People's Republic of China
| | - Wenhong Zhang
- College of Mechanical Engineering, Donghua University, Shanghai 201620, People's Republic of China
| | - Xinyu Li
- Department of Minimally Invasive Comprehensive Treatment of Cancer, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong 250021, People's Republic of China
| | - Jun Chen
- School of Mechanical Engineering, Qilu University of Technology (Shandong Academy of Sciences), Jinan 250353, People's Republic of China
- Shandong Institute of Mechanical Design and Research, Jinan 250353, People's Republic of China
| | - Li Wang
- School of Mechanical Engineering, Qilu University of Technology (Shandong Academy of Sciences), Jinan 250353, People's Republic of China
- Shandong Institute of Mechanical Design and Research, Jinan 250353, People's Republic of China
| |
Collapse
|
32
|
Roegiers I, Gheysens T, Minsart M, De Clercq P, Vanbeversluys K, Rać N, Stroka G, de Croock J, Van de Wiele T, Dubruel P, Arroyo MC. GelMA as scaffold material for epithelial cells to emulate the small intestinal microenvironment. Sci Rep 2025; 15:8214. [PMID: 40064943 PMCID: PMC11893807 DOI: 10.1038/s41598-024-81533-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2024] [Accepted: 11/27/2024] [Indexed: 03/14/2025] Open
Abstract
Host-microbe interactions in the intestine play a significant role in health and disease. Novel scaffolds for host cells, capable of potentially supporting these intricate interactions, are necessary to improve our current systems for mimicking host-microbiota interplay in vitro/ex vivo. In this research paper, we study the application of gelatin methacrylamide (GelMA) as scaffold material for intestinal epithelial cells in terms of permeability, mechanical strength, and biocompatibility. We investigated whether the degree of substitution (DS) of GelMA influences the permeability and found that both high and low DS GelMA show sufficient permeability of biorelevant transport markers. Additionally, we researched epithelial cell adherence and viability, as well as mechanical characteristics of different concentrations of GelMA. All concentrations of hydrogel show long-term biocompatibility for the mono- and co-cultures, despite the goblet-like cells (LS174T) showing lower performance than enterocyte-like cells (Caco-2). The mechanical strength of all hydrogel concentrations was in a physiologically relevant range to be used as scaffold material for intestinal cells. Lastly, we examined the effect of the two sterilization methods, ethylene oxide (EO) and 70% ethanol followed by UVC (EtOH/UVC). We found that the impact of the two methods on the mechanical characteristics was minimal, and we did not find a significant effect between the two methods on cell viability and confluency of Caco-2 cells seeded on the GelMA hydrogels. Based on these results, we conclude that GelMA is a suitable material as a scaffold for intestinal cell types in terms of permeability, mechanical strength and biocompatibility. These findings contribute to the growing field of in vitro modeling of the gut and moves the field further to ensuring more translatable research on host-microbe interactions.
Collapse
Affiliation(s)
- Inez Roegiers
- Center for Microbial Ecology and Technology (CMET), Department of Biotechnology, Faculty of Bioscience Engineering, Ghent University, Ghent, Belgium.
- Polymer Chemistry and Biomaterials Group (PBM), Centre of Macromolecular Chemistry, Department of Organic and Macromolecular Chemistry, Ghent University, Ghent, Belgium.
| | - Tom Gheysens
- Polymer Chemistry and Biomaterials Group (PBM), Centre of Macromolecular Chemistry, Department of Organic and Macromolecular Chemistry, Ghent University, Ghent, Belgium
| | - Manon Minsart
- Polymer Chemistry and Biomaterials Group (PBM), Centre of Macromolecular Chemistry, Department of Organic and Macromolecular Chemistry, Ghent University, Ghent, Belgium
| | - Pieter De Clercq
- Center for Microbial Ecology and Technology (CMET), Department of Biotechnology, Faculty of Bioscience Engineering, Ghent University, Ghent, Belgium
| | - Kim Vanbeversluys
- Center for Microbial Ecology and Technology (CMET), Department of Biotechnology, Faculty of Bioscience Engineering, Ghent University, Ghent, Belgium
| | - Nikoletta Rać
- Center for Microbial Ecology and Technology (CMET), Department of Biotechnology, Faculty of Bioscience Engineering, Ghent University, Ghent, Belgium
| | - Grzegorz Stroka
- Center for Microbial Ecology and Technology (CMET), Department of Biotechnology, Faculty of Bioscience Engineering, Ghent University, Ghent, Belgium
| | - Jana de Croock
- Center for Microbial Ecology and Technology (CMET), Department of Biotechnology, Faculty of Bioscience Engineering, Ghent University, Ghent, Belgium
| | - Tom Van de Wiele
- Center for Microbial Ecology and Technology (CMET), Department of Biotechnology, Faculty of Bioscience Engineering, Ghent University, Ghent, Belgium
| | - Peter Dubruel
- Polymer Chemistry and Biomaterials Group (PBM), Centre of Macromolecular Chemistry, Department of Organic and Macromolecular Chemistry, Ghent University, Ghent, Belgium
| | - Marta Calatayud Arroyo
- Center for Microbial Ecology and Technology (CMET), Department of Biotechnology, Faculty of Bioscience Engineering, Ghent University, Ghent, Belgium
- Department of Food Biotechnology, Institute of Agrochemistry and Food Technology, Spanish National Research Council (CSIC), Valencia, Spain
| |
Collapse
|
33
|
Vanhoeijen R, Okkelman IA, Rogier N, Sedlačík T, Stöbener DD, Devriendt B, Dmitriev RI, Hoogenboom R. Poly(2-alkyl-2-oxazoline) Hydrogels as Synthetic Matrices for Multicellular Spheroid and Intestinal Organoid Cultures. Biomacromolecules 2025; 26:1860-1872. [PMID: 39898884 DOI: 10.1021/acs.biomac.4c01627] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2025]
Abstract
The extracellular matrix (ECM) plays a crucial role in organoid cultures by supporting cell proliferation and differentiation. A key feature of the ECM is its mechanical influence on the surrounding cells, directly affecting their behavior. Matrigel, the most commonly used ECM, is limited by its animal-derived origin, batch variability, and uncontrollable mechanical properties, restricting its use in 3D cell-model-based mechanobiological studies. Poly(2-alkyl-2-oxazoline) (PAOx) synthetic hydrogels represent an appealing alternative because of their reproducibility and versatile chemistry, enabling tuning of hydrogel stiffness and functionalization. Here, we studied PAOx hydrogels with differing compressive moduli for their potential to support 3D cell growth. PAOx hydrogels support spheroid and organoid growth over several days without the addition of ECM components. Furthermore, we discovered intestinal organoid epithelial polarity reversion in PAOx hydrogels and demonstrate how the tunable mechanical properties of PAOx can be used to study effects on the morphology and oxygenation of live multicellular spheroids.
Collapse
Affiliation(s)
- Robin Vanhoeijen
- Supramolecular Chemistry Group, Centre of Macromolecular Chemistry (CMaC), Department of Organic and Macromolecular Chemistry, Faculty of Sciences, Ghent University, Krijgslaan 281-S4, Ghent 9000, Belgium
- Tissue Engineering and Biomaterials Group, Department of Human Structure and Repair, Faculty of Medicine and Health Sciences, Ghent University, C. Heymanslaan 10, Ghent 9000, Belgium
| | - Irina A Okkelman
- Tissue Engineering and Biomaterials Group, Department of Human Structure and Repair, Faculty of Medicine and Health Sciences, Ghent University, C. Heymanslaan 10, Ghent 9000, Belgium
| | - Nette Rogier
- Supramolecular Chemistry Group, Centre of Macromolecular Chemistry (CMaC), Department of Organic and Macromolecular Chemistry, Faculty of Sciences, Ghent University, Krijgslaan 281-S4, Ghent 9000, Belgium
- Tissue Engineering and Biomaterials Group, Department of Human Structure and Repair, Faculty of Medicine and Health Sciences, Ghent University, C. Heymanslaan 10, Ghent 9000, Belgium
| | - Tomáš Sedlačík
- Supramolecular Chemistry Group, Centre of Macromolecular Chemistry (CMaC), Department of Organic and Macromolecular Chemistry, Faculty of Sciences, Ghent University, Krijgslaan 281-S4, Ghent 9000, Belgium
- Hydrogel Lab, Department of Polymers, Faculty of Chemical Technology, University of Chemistry and Technology, Technicka 1903/5, Prague 6 166 28, Czech Republic
| | - Daniel D Stöbener
- Supramolecular Chemistry Group, Centre of Macromolecular Chemistry (CMaC), Department of Organic and Macromolecular Chemistry, Faculty of Sciences, Ghent University, Krijgslaan 281-S4, Ghent 9000, Belgium
| | - Bert Devriendt
- Laboratory of Immunology, Department of Translational Physiology, Infectiology and Public Health, Faculty of Veterinary Medicine, Ghent University, Merelbeke 9820, Belgium
| | - Ruslan I Dmitriev
- Tissue Engineering and Biomaterials Group, Department of Human Structure and Repair, Faculty of Medicine and Health Sciences, Ghent University, C. Heymanslaan 10, Ghent 9000, Belgium
| | - Richard Hoogenboom
- Supramolecular Chemistry Group, Centre of Macromolecular Chemistry (CMaC), Department of Organic and Macromolecular Chemistry, Faculty of Sciences, Ghent University, Krijgslaan 281-S4, Ghent 9000, Belgium
| |
Collapse
|
34
|
Lai W, Geliang H, Bin X, Wang W. Effects of hydrogel stiffness and viscoelasticity on organoid culture: a comprehensive review. Mol Med 2025; 31:83. [PMID: 40033190 PMCID: PMC11877758 DOI: 10.1186/s10020-025-01131-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2024] [Accepted: 02/14/2025] [Indexed: 03/05/2025] Open
Abstract
As an emerging technology, organoids are promising new tools for basic and translational research in disease. Currently, the culture of organoids relies mainly on a type of unknown composition scaffold, namely Matrigel, which may pose problems in studying the effect of mechanical properties on organoids. Hydrogels, a new material with adjustable mechanical properties, can adapt to current studies. In this review, we summarized the synthesis of recent advance in developing definite hydrogel scaffolds for organoid culture and identified the critical parameters for regulating mechanical properties. In addition, classified by different mechanical properties like stiffness and viscoelasticity, we concluded the effect of mechanical properties on the development of organoids and tumor organoids. We hope this review enhances the understanding of the development of organoids by hydrogels and provides more practical approaches to investigating them.
Collapse
Affiliation(s)
- Wei Lai
- Department of Thoracic Surgery, Renmin Hospital of Wuhan University, Wuhan, 430060, China
| | - Hu Geliang
- Department of Orthopedics, Renmin Hospital of Wuhan University, Wuhan, 430060, China
| | - Xu Bin
- Cancer Center, Renmin Hospital of Wuhan University, Wuhan, 430060, China.
| | - Wei Wang
- Department of Thoracic Surgery, Renmin Hospital of Wuhan University, Wuhan, 430060, China.
| |
Collapse
|
35
|
Zhao J, Zhi Y, Ren H, Wang J, Zhao Y. Emerging biotechnologies for engineering liver organoids. Bioact Mater 2025; 45:1-18. [PMID: 39588483 PMCID: PMC11585797 DOI: 10.1016/j.bioactmat.2024.11.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2024] [Revised: 11/02/2024] [Accepted: 11/02/2024] [Indexed: 11/27/2024] Open
Abstract
The engineering construction of the liver has attracted enormous attention. Organoids, as emerging miniature three-dimensional cultivation units, hold significant potential in the biomimetic simulation of liver structure and function. Despite notable successes, organoids still face limitations such as high variability and low maturity. To overcome these challenges, engineering strategies have been established to maintain organoid stability and enhance their efficacy, laying the groundwork for the development of advanced liver organoids. The present review comprehensively summarizes the construction of engineered liver organoids and their prospective applications in biomedicine. Initially, we briefly present the latest research progress on matrix materials that maintain the three-dimensional morphology of organoids. Next, we discuss the manipulative role of engineering technologies in organoid assembly. Additionally, we outline the impact of gene-level regulation on organoid growth and development. Further, we introduce the applications of liver organoids in disease modeling, drug screening and regenerative medicine. Lastly, we overview the current obstacles and forward-looking perspectives on the future of engineered liver organoids. We anticipate that ongoing innovations in engineered liver organoids will lead to significant advancements in medical applications.
Collapse
Affiliation(s)
- Junqi Zhao
- Department of Hepatobiliary Surgery, Hepatobiliary Institute, Nanjing Drum Tower Hospital, Medical School, Nanjing University, Nanjing, 210008, China
- Department of Gastrointestinal Surgery, The First Affiliated Hospital, Wenzhou Medical University, Wenzhou, 325035, China
| | - Yue Zhi
- Department of Hepatobiliary Surgery, Hepatobiliary Institute, Nanjing Drum Tower Hospital, Medical School, Nanjing University, Nanjing, 210008, China
| | - Haozhen Ren
- Department of Hepatobiliary Surgery, Hepatobiliary Institute, Nanjing Drum Tower Hospital, Medical School, Nanjing University, Nanjing, 210008, China
| | - Jinglin Wang
- Department of Hepatobiliary Surgery, Hepatobiliary Institute, Nanjing Drum Tower Hospital, Medical School, Nanjing University, Nanjing, 210008, China
| | - Yuanjin Zhao
- Department of Hepatobiliary Surgery, Hepatobiliary Institute, Nanjing Drum Tower Hospital, Medical School, Nanjing University, Nanjing, 210008, China
- Department of Gastrointestinal Surgery, The First Affiliated Hospital, Wenzhou Medical University, Wenzhou, 325035, China
- Shenzhen Research Institute, Southeast University, Shenzhen, 518038, China
| |
Collapse
|
36
|
Sugihara HY, Okamoto R, Mizutani T. Intestinal organoids: The path towards clinical application. Eur J Cell Biol 2025; 104:151474. [PMID: 39740324 DOI: 10.1016/j.ejcb.2024.151474] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2024] [Revised: 11/14/2024] [Accepted: 11/17/2024] [Indexed: 01/02/2025] Open
Abstract
Organoids have revolutionized the whole field of biology with their ability to model complex three-dimensional human organs in vitro. Intestinal organoids were especially consequential as the first successful long-term culture of intestinal stem cells, which raised hopes for translational medical applications. Despite significant contributions to basic research, challenges remain to develop intestinal organoids into clinical tools for diagnosis, prognosis, and therapy. In this review, we outline the current state of translational research involving adult stem cell and pluripotent stem cell derived intestinal organoids, highlighting the advances and limitations in disease modeling, drug-screening, personalized medicine, and stem cell therapy. Preclinical studies have demonstrated a remarkable functional recapitulation of infectious and genetic diseases, and there is mounting evidence for the reliability of intestinal organoids as a patient-specific avatar. Breakthroughs now allow the generation of structurally and cellularly complex intestinal models to better capture a wider range of intestinal pathophysiology. As the field develops and evolves, there is a need for standardized frameworks for generation, culture, storage, and analysis of intestinal organoids to ensure reproducibility, comparability, and interpretability of these preclinical and clinical studies to ultimately enable clinical translation.
Collapse
Affiliation(s)
- Hady Yuki Sugihara
- Department of Gastroenterology and Hepatology, Institute of Science Tokyo, 1-5-45, Yushima, Bunkyo-ku, Tokyo 113-8510, Japan
| | - Ryuichi Okamoto
- Department of Gastroenterology and Hepatology, Institute of Science Tokyo, 1-5-45, Yushima, Bunkyo-ku, Tokyo 113-8510, Japan
| | - Tomohiro Mizutani
- Department of Gastroenterology and Hepatology, Institute of Science Tokyo, 1-5-45, Yushima, Bunkyo-ku, Tokyo 113-8510, Japan.
| |
Collapse
|
37
|
Cui D, Kong N, Yang W, Yan F. Recent advances in nanoarchitectonics of two-dimensional nanomaterials for dental biosensing and drug delivery. Adv Colloid Interface Sci 2025; 337:103388. [PMID: 39754906 DOI: 10.1016/j.cis.2024.103388] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2024] [Revised: 12/26/2024] [Accepted: 12/27/2024] [Indexed: 01/06/2025]
Abstract
Two-dimensional (2D) nanoarchitectonics involve the creation of functional material assemblies and structures at the nanoscopic level by combining and organizing nanoscale components through various strategies, such as chemical and physical reforming, atomic and molecular manipulation, and self-assembly. Significant advancements have been made in the field, with the goal of producing functional materials from these nanoscale components. 2D nanomaterials, in particular, have gained substantial attention due to their large surface areas which are ideal for numerous surface-active applications. In this review article, nanoarchitectonics of 2D nanomaterials based biomedical applications are discussed. We aim to provide a concise overview of how nanoarchitectonics using 2D nanomaterials can be applied to dental healthcare, with an emphasis on biosensing and drug delivery. By offering a deeper understanding of nanoarchitectonics with programmable structures and predictable properties, we hope to inspire new innovations in the dental bioapplications of 2D nanomaterials.
Collapse
Affiliation(s)
- Di Cui
- Department of Periodontology, Nanjing Stomatological Hospital, Affiliated Hospital of Medical School, Institute of Stomatology, Nanjing University, Nanjing 210008, China; School of Life and Environmental Sciences, Centre for Sustainable Bioproducts, Deakin University Waurn Ponds, Victoria, 3216, Australia
| | - Na Kong
- School of Life and Environmental Sciences, Centre for Sustainable Bioproducts, Deakin University Waurn Ponds, Victoria, 3216, Australia
| | - Wenrong Yang
- School of Life and Environmental Sciences, Centre for Sustainable Bioproducts, Deakin University Waurn Ponds, Victoria, 3216, Australia.
| | - Fuhua Yan
- Department of Periodontology, Nanjing Stomatological Hospital, Affiliated Hospital of Medical School, Institute of Stomatology, Nanjing University, Nanjing 210008, China.
| |
Collapse
|
38
|
Liu Y, Tan J, Zhang N, Qu Z, Li W, Wu Y, Yin H, Liu G, Fu B. Trichinella spiralis excretory/secretory antigens ameliorate porcine epidemic diarrhea virus-induced mucosal damage in porcine intestinal oganoids by alleviating inflammation and promoting tight junction. Int J Parasitol 2025; 55:183-195. [PMID: 39725260 DOI: 10.1016/j.ijpara.2024.12.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2024] [Revised: 10/28/2024] [Accepted: 12/19/2024] [Indexed: 12/28/2024]
Abstract
Trichinella spiralis and porcine epidemic diarrhea virus (PEDV) are two infectious swine pathogens. Parasite excretory/secretory antigens play critical roles in various disease processes. To explore the coexistence mechanism of two pathogens infecting the same host, the intestinal organoid was utilized to reproduce these biological processes. In this study, we investigated the effects of T. spiralis excretory/secretory antigens (TsES) on PEDV-induced inflammatory regulation, lesion recovery, and mucosal barrier repair in porcine intestinal organoids. The results showed that PEDV effectively infected the porcine intestinal organoids. Next, TsES inhibited pro-inflammatory cytokines and increased the anti-inflammatory cytokines produced by PEDV-infected porcine intestinal organoids. Further, four-dimensional (4D) label-free quantitative proteomics and western blotting confirmed that TsES regulate the inflammation caused by PEDV infection through the nuclear factor kappa-B (NF-κB) pathway. In addition, TsES promoted cell proliferation, inhibited apoptosis, and reduced PEDV-induced lesions in intestinal organoids. The elevated secretory immunoglobulin A (sIgA) levels caused by PEDV infection were downregulated by TsES treatment in intestinal organoids. TsES treatment reversed the mucosal barrier damage caused by PEDV infection in intestinal organoids. Finally, PEDV replication increased after TsES treatment in organoids. We highlight the potential of TsES to ameliorate PEDV-induced inflammation, mucosal lesions, and barrier damage in porcine intestinal organoids. TsES also contribute to PEDV replication. This study presents a novel research model for research on host-virus-parasite interactions, while also providing a theoretical foundation to consider parasite derivatives as a potential adjunctive therapy for intestinal inflammation.
Collapse
Affiliation(s)
- Yinju Liu
- State Key Laboratory for Animal Disease Control and Prevention, Key Laboratory of Veterinary Parasitology of Gansu Province, Gansu Province Research Center for Basic Disciplines of Pathogen Biology, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou 730046, China
| | - Jinlong Tan
- State Key Laboratory for Animal Disease Control and Prevention, Key Laboratory of Veterinary Parasitology of Gansu Province, Gansu Province Research Center for Basic Disciplines of Pathogen Biology, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou 730046, China; Institute of Traditional Chinese Medicine Health Industry, China Academy of Chinese Medical Sciences, Nanchang 330115, China
| | - Nianzhang Zhang
- State Key Laboratory for Animal Disease Control and Prevention, Key Laboratory of Veterinary Parasitology of Gansu Province, Gansu Province Research Center for Basic Disciplines of Pathogen Biology, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou 730046, China.
| | - Zigang Qu
- State Key Laboratory for Animal Disease Control and Prevention, Key Laboratory of Veterinary Parasitology of Gansu Province, Gansu Province Research Center for Basic Disciplines of Pathogen Biology, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou 730046, China
| | - Wenhui Li
- State Key Laboratory for Animal Disease Control and Prevention, Key Laboratory of Veterinary Parasitology of Gansu Province, Gansu Province Research Center for Basic Disciplines of Pathogen Biology, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou 730046, China
| | - Yaodong Wu
- State Key Laboratory for Animal Disease Control and Prevention, Key Laboratory of Veterinary Parasitology of Gansu Province, Gansu Province Research Center for Basic Disciplines of Pathogen Biology, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou 730046, China
| | - Hong Yin
- State Key Laboratory for Animal Disease Control and Prevention, Key Laboratory of Veterinary Parasitology of Gansu Province, Gansu Province Research Center for Basic Disciplines of Pathogen Biology, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou 730046, China; Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Disease, Yangzhou 225009, China
| | - Guangliang Liu
- State Key Laboratory for Animal Disease Control and Prevention, Key Laboratory of Veterinary Parasitology of Gansu Province, Gansu Province Research Center for Basic Disciplines of Pathogen Biology, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou 730046, China
| | - Baoquan Fu
- State Key Laboratory for Animal Disease Control and Prevention, Key Laboratory of Veterinary Parasitology of Gansu Province, Gansu Province Research Center for Basic Disciplines of Pathogen Biology, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou 730046, China; Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Disease, Yangzhou 225009, China.
| |
Collapse
|
39
|
Artegiani B, Hendriks D. Organoids from pluripotent stem cells and human tissues: When two cultures meet each other. Dev Cell 2025; 60:493-511. [PMID: 39999776 DOI: 10.1016/j.devcel.2025.01.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2024] [Revised: 06/13/2024] [Accepted: 01/10/2025] [Indexed: 02/27/2025]
Abstract
Human organoids are a widely used tool in cell biology to study homeostatic processes, disease, and development. The term organoids covers a plethora of model systems from different cellular origins that each have unique features and applications but bring their own challenges. This review discusses the basic principles underlying organoids generated from pluripotent stem cells (PSCs) as well as those derived from tissue stem cells (TSCs). We consider how well PSC- and TSC-organoids mimic the different intended organs in terms of cellular complexity, maturity, functionality, and the ongoing efforts to constitute predictive complex models of in vivo situations. We discuss the advantages and limitations associated with each system to answer different biological questions including in the field of cancer and developmental biology, and with respect to implementing emerging advanced technologies, such as (spatial) -omics analyses, CRISPR screens, and high-content imaging screens. We postulate how the two fields may move forward together, integrating advantages of one to the other.
Collapse
Affiliation(s)
| | - Delilah Hendriks
- Princess Máxima Center for Pediatric Oncology, Utrecht, the Netherlands.
| |
Collapse
|
40
|
Wu H, Feng E, Yin H, Zhang Y, Chen G, Zhu B, Yue X, Zhang H, Liu Q, Xiong L. Biomaterials for neuroengineering: applications and challenges. Regen Biomater 2025; 12:rbae137. [PMID: 40007617 PMCID: PMC11855295 DOI: 10.1093/rb/rbae137] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2024] [Revised: 10/19/2024] [Accepted: 11/03/2024] [Indexed: 02/27/2025] Open
Abstract
Neurological injuries and diseases are a leading cause of disability worldwide, underscoring the urgent need for effective therapies. Neural regaining and enhancement therapies are seen as the most promising strategies for restoring neural function, offering hope for individuals affected by these conditions. Despite their promise, the path from animal research to clinical application is fraught with challenges. Neuroengineering, particularly through the use of biomaterials, has emerged as a key field that is paving the way for innovative solutions to these challenges. It seeks to understand and treat neurological disorders, unravel the nature of consciousness, and explore the mechanisms of memory and the brain's relationship with behavior, offering solutions for neural tissue engineering, neural interfaces and targeted drug delivery systems. These biomaterials, including both natural and synthetic types, are designed to replicate the cellular environment of the brain, thereby facilitating neural repair. This review aims to provide a comprehensive overview for biomaterials in neuroengineering, highlighting their application in neural functional regaining and enhancement across both basic research and clinical practice. It covers recent developments in biomaterial-based products, including 2D to 3D bioprinted scaffolds for cell and organoid culture, brain-on-a-chip systems, biomimetic electrodes and brain-computer interfaces. It also explores artificial synapses and neural networks, discussing their applications in modeling neural microenvironments for repair and regeneration, neural modulation and manipulation and the integration of traditional Chinese medicine. This review serves as a comprehensive guide to the role of biomaterials in advancing neuroengineering solutions, providing insights into the ongoing efforts to bridge the gap between innovation and clinical application.
Collapse
Affiliation(s)
- Huanghui Wu
- Translational Research Institute of Brain and Brain-Like Intelligence, Shanghai Key Laboratory of Anesthesiology and Brain Functional Modulation, Clinical Research Center for Anesthesiology and Perioperative Medicine, Department of Anesthesiology and Perioperative Medicine, Shanghai Fourth People’s Hospital, School of Medicine, Tongji University, Shanghai 200434, China
| | - Enduo Feng
- Translational Research Institute of Brain and Brain-Like Intelligence, Shanghai Key Laboratory of Anesthesiology and Brain Functional Modulation, Clinical Research Center for Anesthesiology and Perioperative Medicine, Department of Anesthesiology and Perioperative Medicine, Shanghai Fourth People’s Hospital, School of Medicine, Tongji University, Shanghai 200434, China
| | - Huanxin Yin
- Translational Research Institute of Brain and Brain-Like Intelligence, Shanghai Key Laboratory of Anesthesiology and Brain Functional Modulation, Clinical Research Center for Anesthesiology and Perioperative Medicine, Department of Anesthesiology and Perioperative Medicine, Shanghai Fourth People’s Hospital, School of Medicine, Tongji University, Shanghai 200434, China
| | - Yuxin Zhang
- Translational Research Institute of Brain and Brain-Like Intelligence, Shanghai Key Laboratory of Anesthesiology and Brain Functional Modulation, Clinical Research Center for Anesthesiology and Perioperative Medicine, Department of Anesthesiology and Perioperative Medicine, Shanghai Fourth People’s Hospital, School of Medicine, Tongji University, Shanghai 200434, China
| | - Guozhong Chen
- Translational Research Institute of Brain and Brain-Like Intelligence, Shanghai Key Laboratory of Anesthesiology and Brain Functional Modulation, Clinical Research Center for Anesthesiology and Perioperative Medicine, Department of Anesthesiology and Perioperative Medicine, Shanghai Fourth People’s Hospital, School of Medicine, Tongji University, Shanghai 200434, China
| | - Beier Zhu
- Translational Research Institute of Brain and Brain-Like Intelligence, Shanghai Key Laboratory of Anesthesiology and Brain Functional Modulation, Clinical Research Center for Anesthesiology and Perioperative Medicine, Department of Anesthesiology and Perioperative Medicine, Shanghai Fourth People’s Hospital, School of Medicine, Tongji University, Shanghai 200434, China
| | - Xuezheng Yue
- School of Materials and Chemistry, University of Shanghai for Science and Technology, Shanghai 200093, China
| | - Haiguang Zhang
- Rapid Manufacturing Engineering Center, School of Mechatronical Engineering and Automation, Shanghai University, Shanghai 200444, China
- Shanghai Key Laboratory of Intelligent Manufacturing and Robotics, Shanghai University, Shanghai 200072, China
| | - Qiong Liu
- Translational Research Institute of Brain and Brain-Like Intelligence, Shanghai Key Laboratory of Anesthesiology and Brain Functional Modulation, Clinical Research Center for Anesthesiology and Perioperative Medicine, Department of Anesthesiology and Perioperative Medicine, Shanghai Fourth People’s Hospital, School of Medicine, Tongji University, Shanghai 200434, China
- State Key Laboratory of Molecular Engineering of Polymers, Department of Macromolecular Science, Fudan University, Shanghai 200438, China
| | - Lize Xiong
- Translational Research Institute of Brain and Brain-Like Intelligence, Shanghai Key Laboratory of Anesthesiology and Brain Functional Modulation, Clinical Research Center for Anesthesiology and Perioperative Medicine, Department of Anesthesiology and Perioperative Medicine, Shanghai Fourth People’s Hospital, School of Medicine, Tongji University, Shanghai 200434, China
| |
Collapse
|
41
|
Sheehan A, Okkelman IA, Groslambert G, Bucher C, Dmitriev RI, Filatov MA. Optoelectronic Properties and Fluorescence Lifetime Imaging Application of Donor-Acceptor Dyads Derived From 2,6-DicarboxyBODIPY. Chemistry 2025; 31:e202404188. [PMID: 39740050 DOI: 10.1002/chem.202404188] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2024] [Revised: 12/12/2024] [Accepted: 12/31/2024] [Indexed: 01/02/2025]
Abstract
Donor-acceptor BODIPY dyads, functionalized at the 2 and 6 positions with benzyl ester (BDP-DE) or carboxylic acid (BDP-DA) groups, were synthesized, and their optoelectronic properties were investigated. Carbonyl groups were found to increase the reduction potential of the BODIPY core by 0.15-0.4 eV compared to regular alkyl-substituted BODIPYs. These compounds exhibited efficient intramolecular charge separation and triplet state formation through the spin-orbit charge transfer intersystem crossing (SOCT-ISC) process, achieving singlet oxygen quantum yields of up to 92 %, depending on the solvent polarity. Notably, the fluorescence and singlet oxygen generation of BDP-DAs were found to depend on the ionization state of the carboxylic groups. Time-resolved fluorescence measurements revealed that complexation of BDP-DAs with bovine serum albumine (BSA) significantly extended their excited state lifetimes. Fluorescence lifetime imaging microscopy (FLIM) studies of human colorectal carcinoma (HCT116) cells and pig small intestinal organoids (enteroids) provided insights into subcellular localization. The diacid with 2,4-dimethoxyphenyl group at the meso-position (DA1) displayed longer lifetimes in lipid-droplet-like structures and shorter lifetimes in cytoplasmic regions. The diacid containing a meso-anthracenyl group (DA2) formed 'islands' in cell monolayers, exhibiting a distinct lifetime gradient from the periphery to the center. These results highlight the potential of donor-acceptor BODIPYs as fluorescent probes for biological imaging, particularly in revealing subtle differences in cellular environments.
Collapse
Affiliation(s)
- Aimee Sheehan
- School of Chemical and Biopharmaceutical Sciences, Technological University Dublin, City Campus, Grangegorman, Dublin 7, Ireland
| | - Irina A Okkelman
- Tissue Engineering and Biomaterials Group, Department of Human Structure and Repair, Faculty of Medicine and Health Sciences, Ghent University, The Core, C. Heymanslaan 10, 9000, Ghent, Belgium
| | - Geoffrey Groslambert
- CNRS, ENS de Lyon, LCH, UMR 5182, 46 Allée d'Italie, 69342, Lyon Cedex 07, France
| | - Christophe Bucher
- CNRS, ENS de Lyon, LCH, UMR 5182, 46 Allée d'Italie, 69342, Lyon Cedex 07, France
| | - Ruslan I Dmitriev
- Tissue Engineering and Biomaterials Group, Department of Human Structure and Repair, Faculty of Medicine and Health Sciences, Ghent University, The Core, C. Heymanslaan 10, 9000, Ghent, Belgium
| | - Mikhail A Filatov
- School of Chemical and Biopharmaceutical Sciences, Technological University Dublin, City Campus, Grangegorman, Dublin 7, Ireland
| |
Collapse
|
42
|
Günther D, Bergerbit C, Marsee A, Vedaraman S, Pueyo Moliner A, Bastard C, Eelen G, Gerardo Nava JL, Dewerchin M, Carmeliet P, Kramann R, Schneeberger K, Spee B, De Laporte L. Synergizing bioprinting and 3D cell culture to enhance tissue formation in printed synthetic constructs. Biofabrication 2025; 17:025015. [PMID: 39854847 DOI: 10.1088/1758-5090/adae37] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2024] [Accepted: 01/24/2025] [Indexed: 01/27/2025]
Abstract
Bioprinting is currently the most promising method to biofabricate complex tissuesin vitrowith the potential to transform the future of organ transplantation and drug discovery. Efforts to create such tissues are, however, almost exclusively based on animal-derived materials, such as gelatin methacryloyl, which have demonstrated efficacy in bioprinting of complex tissues. While these materials are already used in clinical applications, uncertainty about their safety still remains due to their animal origin. Alternatively, synthetic bioinks have been developed that match the printability of natural bioinks but lack their biological complexity, and thereby often fail to support cell growth and facilitate tissue formation. Additionally, most synthetic materials do not meet the mechanical demands of bioprint stable constructs while providing a suitable environment for cells to grow, limiting the number of available bioinks. To bridge this gap and synergize bioprinting and 3D cell culture, we developed a polyethylene glycol-based bioink system to promote the growth and spreading of cell spheroids that consist of human primary endothelial cells and fibroblasts. The 3D bioprinted centimeter-scale constructs have a high shape fidelity and accelerated softening to provide sufficient space for cells to grow. Adjusting the rate of degradability, induced by the integration of ester-functionalized crosslinkers in addition to protease cleavable crosslinkers into the hydrogel network, improves the growth of spheroids in larger printed hydrogel constructs containing an interconnected channel structure. The perfusable constructs enable extensive spheroid sprouting and the formation of a cellular network upon fusion of sprouts as initial steps toward tissue formation with the potential for clinical translation.
Collapse
Affiliation(s)
- Daniel Günther
- DWI-Leibniz Institute for Interactive Materials, Aachen, Germany
- Institute of Technical and Macromolecular Chemistry, Advanced Materials for Biomedicine (AMB), RWTH Aachen University, Aachen, Germany
- Institute of Applied Medical Engineering, AMB, RWTH Aachen University Hospital, Center for Biohybrid Medical Systems, Aachen, Germany
| | - Cédric Bergerbit
- DWI-Leibniz Institute for Interactive Materials, Aachen, Germany
- Institute of Technical and Macromolecular Chemistry, Advanced Materials for Biomedicine (AMB), RWTH Aachen University, Aachen, Germany
- Institute of Applied Medical Engineering, AMB, RWTH Aachen University Hospital, Center for Biohybrid Medical Systems, Aachen, Germany
| | - Ary Marsee
- Department of Clinical Sciences, Faculty of Veterinary Medicine, Utrecht University, Utrecht, The Netherlands
| | - Sitara Vedaraman
- DWI-Leibniz Institute for Interactive Materials, Aachen, Germany
- Institute of Technical and Macromolecular Chemistry, Advanced Materials for Biomedicine (AMB), RWTH Aachen University, Aachen, Germany
- Institute of Applied Medical Engineering, AMB, RWTH Aachen University Hospital, Center for Biohybrid Medical Systems, Aachen, Germany
| | - Alba Pueyo Moliner
- Department of Clinical Sciences, Faculty of Veterinary Medicine, Utrecht University, Utrecht, The Netherlands
| | - Céline Bastard
- DWI-Leibniz Institute for Interactive Materials, Aachen, Germany
- Institute of Technical and Macromolecular Chemistry, Advanced Materials for Biomedicine (AMB), RWTH Aachen University, Aachen, Germany
- Institute of Applied Medical Engineering, AMB, RWTH Aachen University Hospital, Center for Biohybrid Medical Systems, Aachen, Germany
| | - Guy Eelen
- Laboratory of Angiogenesis and Vascular Metabolism, Department of Oncology and Leuven Cancer Institute (LKI), KU Leuven, and VIB Center for Cancer Biology, Leuven, Belgium
| | - José Luis Gerardo Nava
- DWI-Leibniz Institute for Interactive Materials, Aachen, Germany
- Institute of Applied Medical Engineering, AMB, RWTH Aachen University Hospital, Center for Biohybrid Medical Systems, Aachen, Germany
| | - Mieke Dewerchin
- Laboratory of Angiogenesis and Vascular Metabolism, Department of Oncology and Leuven Cancer Institute (LKI), KU Leuven, and VIB Center for Cancer Biology, Leuven, Belgium
| | - Peter Carmeliet
- Laboratory of Angiogenesis and Vascular Metabolism, Department of Oncology and Leuven Cancer Institute (LKI), KU Leuven, and VIB Center for Cancer Biology, Leuven, Belgium
| | - Rafael Kramann
- Department of Nephrology and Clinical Immunology, Medical Faculty, RWTH Aachen University, Aachen, Germany
- Department of Internal Medicine, Nephrology, and Transplantation, Erasmus Medical Center, Rotterdam, The Netherlands
| | - Kerstin Schneeberger
- Department of Clinical Sciences, Faculty of Veterinary Medicine, Utrecht University, Utrecht, The Netherlands
| | - Bart Spee
- Department of Clinical Sciences, Faculty of Veterinary Medicine, Utrecht University, Utrecht, The Netherlands
| | - Laura De Laporte
- DWI-Leibniz Institute for Interactive Materials, Aachen, Germany
- Institute of Technical and Macromolecular Chemistry, Advanced Materials for Biomedicine (AMB), RWTH Aachen University, Aachen, Germany
- Institute of Applied Medical Engineering, AMB, RWTH Aachen University Hospital, Center for Biohybrid Medical Systems, Aachen, Germany
| |
Collapse
|
43
|
Wu H, Wang J, Liu S, Wang Y, Tang X, Xie J, Wang N, Shan H, Chen S, Zhang X, Zeng W, Chen C, Fu Y, Lai L, Duan Y. Large-Scale Production of Expandable Hepatoblast Organoids and Polarised Hepatocyte Organoids From hESCs Under 3D Static and Dynamic Suspension Conditions. Cell Prolif 2025:e70001. [PMID: 39921573 DOI: 10.1111/cpr.70001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2024] [Revised: 01/17/2025] [Accepted: 01/27/2025] [Indexed: 02/10/2025] Open
Abstract
To date, generating viable and functional hepatocytes in large scale remains challenge. By employing 3D suspension condition with the support of low concentration Matrigel, a novel culture system was developed to generate expandable hepatoblast organoids (HB-orgs) and mature polarised hepatocyte organoids (P-hep-orgs) from human embryonic stem cells (hESCs) in both dishes and bioreactors. scRNA-seq and functional assays were used to characterise HB-orgs and P-hep-orgs. hESC-derived HB-orgs could proliferate at least for 15 passages, leading to 1012 in total cells in 4 weeks. P-hep-orgs differentiated from HB-orgs displayed characteristics of mature hepatocytes with polarisation. Moreover, single-cell RNA sequencing exhibited that over 40% of cells in P-hep-orgs were highly fidelity with human primary hepatocytes. Eventually, large-scale production of P-hep-orgs could be generated from massively expanded HB-orgs within 1 week with similar number in bioreactors, which were achieved by the enhancements in energy metabolism contribute to the expansion of HB-orgs and maturation of P-hep-orgs in bioreactors. By providing a cost-efficient and robust platform, our study represents a significant step toward manufacturing large-scale functioning hESC-derived hepatocytes for cell-based therapeutics, disease modelling, pharmacology and toxicology studies.
Collapse
Affiliation(s)
- Haibin Wu
- Laboratory of Stem Cells and Translational Medicine, Institute for Medical Research, The Second Affiliated Hospital, School of Medicine, South China University of Technology, Guangzhou, China
- Laboratory of Stem Cells and Translational Medicine, Institute for Life Science, School of Medicine, South China University of Technology, Guangzhou, China
| | - Jue Wang
- Laboratory of Stem Cells and Translational Medicine, Institute for Life Science, School of Medicine, South China University of Technology, Guangzhou, China
| | - Shoupei Liu
- Laboratory of Stem Cells and Translational Medicine, Institute for Medical Research, The Second Affiliated Hospital, School of Medicine, South China University of Technology, Guangzhou, China
- Laboratory of Stem Cells and Translational Medicine, Institute for Life Science, School of Medicine, South China University of Technology, Guangzhou, China
| | - Yiyu Wang
- Laboratory of Stem Cells and Translational Medicine, Institute for Medical Research, The Second Affiliated Hospital, School of Medicine, South China University of Technology, Guangzhou, China
- Laboratory of Stem Cells and Translational Medicine, Institute for Life Science, School of Medicine, South China University of Technology, Guangzhou, China
| | - Xianglian Tang
- Laboratory of Stem Cells and Translational Medicine, Institute for Life Science, School of Medicine, South China University of Technology, Guangzhou, China
| | - Jinghe Xie
- Laboratory of Stem Cells and Translational Medicine, Institute for Life Science, School of Medicine, South China University of Technology, Guangzhou, China
- School of Biomedical Sciences and Engineering, South China University of Technology, Guangzhou International Campus, Guangzhou, China
| | - Ning Wang
- Laboratory of Stem Cells and Translational Medicine, Institute for Medical Research, The Second Affiliated Hospital, School of Medicine, South China University of Technology, Guangzhou, China
- Laboratory of Stem Cells and Translational Medicine, Institute for Life Science, School of Medicine, South China University of Technology, Guangzhou, China
| | - Huanhuan Shan
- Laboratory of Stem Cells and Translational Medicine, Institute for Medical Research, The Second Affiliated Hospital, School of Medicine, South China University of Technology, Guangzhou, China
- Laboratory of Stem Cells and Translational Medicine, Institute for Life Science, School of Medicine, South China University of Technology, Guangzhou, China
| | - Sen Chen
- Laboratory of Stem Cells and Translational Medicine, Institute for Medical Research, The Second Affiliated Hospital, School of Medicine, South China University of Technology, Guangzhou, China
- Laboratory of Stem Cells and Translational Medicine, Institute for Life Science, School of Medicine, South China University of Technology, Guangzhou, China
| | - Xueyan Zhang
- Laboratory of Stem Cells and Translational Medicine, Institute for Medical Research, The Second Affiliated Hospital, School of Medicine, South China University of Technology, Guangzhou, China
- Laboratory of Stem Cells and Translational Medicine, Institute for Life Science, School of Medicine, South China University of Technology, Guangzhou, China
| | - Weiping Zeng
- Laboratory of Stem Cells and Translational Medicine, Institute for Life Science, School of Medicine, South China University of Technology, Guangzhou, China
| | - Chuxin Chen
- Laboratory of Stem Cells and Translational Medicine, Institute for Medical Research, The Second Affiliated Hospital, School of Medicine, South China University of Technology, Guangzhou, China
- Laboratory of Stem Cells and Translational Medicine, Institute for Life Science, School of Medicine, South China University of Technology, Guangzhou, China
| | - Yinjie Fu
- Laboratory of Stem Cells and Translational Medicine, Institute for Medical Research, The Second Affiliated Hospital, School of Medicine, South China University of Technology, Guangzhou, China
- Laboratory of Stem Cells and Translational Medicine, Institute for Life Science, School of Medicine, South China University of Technology, Guangzhou, China
| | - Liangxue Lai
- Key Laboratory of Regenerative Biology, South China Institute for Stem Cell, Biology and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China
| | - Yuyou Duan
- Laboratory of Stem Cells and Translational Medicine, Institute for Medical Research, The Second Affiliated Hospital, School of Medicine, South China University of Technology, Guangzhou, China
- Laboratory of Stem Cells and Translational Medicine, Institute for Life Science, School of Medicine, South China University of Technology, Guangzhou, China
- National Engineering Research Center for Tissue Restoration and Reconstruction, South China University of Technology, Guangzhou, China
- The Innovation Centre of Ministry of Education for Development and Diseases, The Second Affiliated Hospital, School of Medicine, South China University of Technology, Guangzhou, China
| |
Collapse
|
44
|
Shao Y, Wang J, Jin A, Jiang S, Lei L, Liu L. Biomaterial-assisted organoid technology for disease modeling and drug screening. Mater Today Bio 2025; 30:101438. [PMID: 39866785 PMCID: PMC11757232 DOI: 10.1016/j.mtbio.2024.101438] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2024] [Revised: 12/10/2024] [Accepted: 12/30/2024] [Indexed: 01/12/2025] Open
Abstract
Developing disease models and screening for effective drugs are key areas of modern medical research. Traditional methodologies frequently fall short in precisely replicating the intricate architecture of bodily tissues and organs. Nevertheless, recent advancements in biomaterial-assisted organoid technology have ushered in a paradigm shift in biomedical research. This innovative approach enables the cultivation of three-dimensional cellular structures in vitro that closely emulate the structural and functional attributes of organs, offering physiologically superior models compared to conventional techniques. The evolution of biomaterials plays a pivotal role in supporting the culture and development of organ tissues, thereby facilitating more accurate disease state modeling and the rigorous evaluation of drug efficacy and safety profiles. In this review, we will explore the roles that various biomaterials play in organoid development, examine the fundamental principles and advantages of utilizing these technologies in constructing disease models, and highlight recent advances and practical applications in drug screening using disease-specific organoid models. Additionally, the challenges and future directions of organoid technology are discussed. Through continued research and innovation, we aim to make remarkable strides in disease treatment and drug development, ultimately enhancing patient quality of life.
Collapse
Affiliation(s)
- Yunyuan Shao
- Key Laboratory of Artificial Organs and Computational Medicine in Zhejiang Province, Institute of Translational Medicine, Zhejiang Shuren University, Hangzhou, 310015, China
| | - Juncheng Wang
- The Third Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325200, China
| | - Anqi Jin
- Key Laboratory of Artificial Organs and Computational Medicine in Zhejiang Province, Institute of Translational Medicine, Zhejiang Shuren University, Hangzhou, 310015, China
| | - Shicui Jiang
- The Third Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325200, China
| | - Lanjie Lei
- Key Laboratory of Artificial Organs and Computational Medicine in Zhejiang Province, Institute of Translational Medicine, Zhejiang Shuren University, Hangzhou, 310015, China
| | - Liangle Liu
- The Third Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325200, China
| |
Collapse
|
45
|
Verstegen MMA, Coppes RP, Beghin A, De Coppi P, Gerli MFM, de Graeff N, Pan Q, Saito Y, Shi S, Zadpoor AA, van der Laan LJW. Clinical applications of human organoids. Nat Med 2025; 31:409-421. [PMID: 39901045 DOI: 10.1038/s41591-024-03489-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2024] [Accepted: 12/17/2024] [Indexed: 02/05/2025]
Abstract
Organoids are innovative three-dimensional and self-organizing cell cultures of various lineages that can be used to study diverse tissues and organs. Human organoids have dramatically increased our understanding of developmental and disease biology. They provide a patient-specific model to study known diseases, with advantages over animal models, and can also provide insights into emerging and future health threats related to climate change, zoonotic infections, environmental pollutants or even microgravity during space exploration. Furthermore, organoids show potential for regenerative cell therapies and organ transplantation. Still, several challenges for broad clinical application remain, including inefficiencies in initiation and expansion, increasing model complexity and difficulties with upscaling clinical-grade cultures and developing more organ-specific human tissue microenvironments. To achieve the full potential of organoid technology, interdisciplinary efforts are needed, integrating advances from biology, bioengineering, computational science, ethics and clinical research. In this Review, we showcase pivotal achievements in epithelial organoid research and technologies and provide an outlook for the future of organoids in advancing human health and medicine.
Collapse
Affiliation(s)
- Monique M A Verstegen
- Department of Surgery, Erasmus MC Transplant Institute, University Medical Center Rotterdam, Rotterdam, the Netherlands.
| | - Rob P Coppes
- Departments of Biomedical Sciences and Radiation Oncology, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
| | - Anne Beghin
- Mechanobiology Institute, National University of Singapore, Singapore, Singapore
- Department of Microbiology and Immunology, Immunology Translational Research Program, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
- Centre for Research and Engineering in Space Technology, Universite Libre de Bruxelles, Bruxelles, Belgium
| | - Paolo De Coppi
- Stem Cell and Regenerative Medicine Section, Zayed Centre for Research into Rare Disease in Children, Great Ormond Street Institute of Child Health, University College London, London, UK
| | - Mattia F M Gerli
- Division of Surgery and Interventional Science, Department of Surgical Biotechnology, University College London, London, UK
| | - Nienke de Graeff
- Department of Medical Ethics and Health Law, Leiden University Medical Center, Leiden University, Leiden, the Netherlands
- The Novo Nordisk Foundation Center for Stem Cell Medicine (reNEW), Leiden Node, Leiden, the Netherlands
| | - Qiuwei Pan
- Department of Gastroenterology and Hepatology, Erasmus MC-University Medical Center, Rotterdam, the Netherlands
| | - Yoshimasa Saito
- Division of Pharmacotherapeutics, Keio University Faculty of Pharmacy, Tokyo, Japan
| | - Shaojun Shi
- Department of Organ Transplantation, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China
| | - Amir A Zadpoor
- Department of Biomechanical Engineering, Faculty of Mechanical, Maritime, and Materials Engineering, Delft University of Technology (TU Delft), Delft, the Netherlands
| | - Luc J W van der Laan
- Department of Surgery, Erasmus MC Transplant Institute, University Medical Center Rotterdam, Rotterdam, the Netherlands
- Department of Biomechanical Engineering, Faculty of Mechanical, Maritime, and Materials Engineering, Delft University of Technology (TU Delft), Delft, the Netherlands
| |
Collapse
|
46
|
Zhao Z, Wu X, Zhang T, Zhou M, Liu S, Yang R, Li JP. Evaluation of Multispecific Drugs Based on Patient-Derived Immunocompetent Tumor Organoids. Chembiochem 2025; 26:e202400731. [PMID: 39800663 DOI: 10.1002/cbic.202400731] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2024] [Revised: 12/12/2024] [Indexed: 01/24/2025]
Abstract
The evolution of antitumor drug development has transitioned from single-agent chemotherapy to targeted therapy, immunotherapy, and more recently, multispecific drugs. These innovative drugs target multiple cellular or molecular pathways simultaneously, offering a more comprehensive anticancer approach and addressing some of the limitations inherent in traditional monotherapies. However, preclinical assessment of multispecific drugs remains challenging, as conventional tumor models often lack the necessary complexity to accurately reflect the interactions between various cell types and targets. Patient-derived immunocompetent tumor organoids (PDITOs), which incorporate both tumor cells and immune cells, present a promising platform for the evaluation of these drugs. Beyond their use in drug evaluation, PDITOs can also be utilized in personalized drug screening and predicting patient-specific treatment outcomes, thus advancing both multispecific drug development and precision medicine. This perspective discusses the current landscape of multispecific drug development and the methodologies for constructing PDITOs. It also addresses the associated challenges and introduces the concept of employing these organoids to optimize the evaluation and rational design of multispecific drug therapies.
Collapse
Affiliation(s)
- Zihan Zhao
- Department of Urology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, 321 Zhongshan Rd, Nanjing, Jiangsu, 210008, China
- State Key Laboratory of Coordination Chemistry, Chemistry and Biomedicine Innovation Center (ChemBIC), School of Chemistry and Chemical Engineering Nanjing University, 163 Xianlin Avenue, Nanjing, Jiangsu, 210023, China
| | - Xiangyu Wu
- Department of Urology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, 321 Zhongshan Rd, Nanjing, Jiangsu, 210008, China
| | - Tianyang Zhang
- State Key Laboratory of Coordination Chemistry, Chemistry and Biomedicine Innovation Center (ChemBIC), School of Chemistry and Chemical Engineering Nanjing University, 163 Xianlin Avenue, Nanjing, Jiangsu, 210023, China
- University of Glasgow, University Avenue, Glasgow, G12 8QQ, UK
| | - Meng Zhou
- Department of Urology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, 321 Zhongshan Rd, Nanjing, Jiangsu, 210008, China
| | - Siyang Liu
- Department of Urology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, 321 Zhongshan Rd, Nanjing, Jiangsu, 210008, China
| | - Rong Yang
- Department of Urology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, 321 Zhongshan Rd, Nanjing, Jiangsu, 210008, China
| | - Jie P Li
- State Key Laboratory of Coordination Chemistry, Chemistry and Biomedicine Innovation Center (ChemBIC), School of Chemistry and Chemical Engineering Nanjing University, 163 Xianlin Avenue, Nanjing, Jiangsu, 210023, China
| |
Collapse
|
47
|
Aksoy SA, Earl J, Grahovac J, Karakas D, Lencioni G, Sığırlı S, Bijlsma MF. Organoids, tissue slices and organotypic cultures: Advancing our understanding of pancreatic ductal adenocarcinoma through in vitro and ex vivo models. Semin Cancer Biol 2025; 109:10-24. [PMID: 39730107 DOI: 10.1016/j.semcancer.2024.12.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2024] [Revised: 12/14/2024] [Accepted: 12/19/2024] [Indexed: 12/29/2024]
Abstract
Pancreatic ductal adenocarcinoma (PDAC) has one of the worst prognoses of all common solid cancers. For the large majority of PDAC patients, only systemic therapies with very limited efficacy are indicated. In addition, immunotherapies have not brought the advances seen in other cancer types. Several key characteristics of PDAC contribute to poor treatment outcomes, and in this review, we will discuss how these characteristics are best captured in currently available ex vivo or in vitro model systems. For instance, PDAC is hallmarked by a highly desmoplastic and immune-suppressed tumor microenvironment that impacts disease progression and therapy resistance. Also, large differences in tumor biology exist between and within tumors, complicating treatment decisions. Furthermore, PDAC has a very high propensity for locally invasive and metastatic growth. The use of animal models is often not desirable or feasible and several in vitro and ex vivo model systems have been developed, such as organotypic cocultures and tissue slices, among others. However, the absence of a full host organism impacts the ability of these models to accurately capture the characteristics that contribute to poor outcomes in PDAC. We will discuss the caveats and advantages of these model systems in the context of PDAC's key characteristics and provide recommendations on model choice and the possibilities for optimization. These considerations should be of use to researchers aiming to study PDAC in the in vitro setting.
Collapse
Affiliation(s)
- Secil Ak Aksoy
- Bursa Uludag University, Faculty of Medicine, Department of Medical Microbiology, Bursa, Turkey
| | - Julie Earl
- Ramón y Cajal Health Research Institute (IRYCIS), Biomodels and Biomodels Platform Hospital Ramón y Cajal-IRYCIS, Carretera Colmenar Km 9,100, Madrid 28034, Spain; The Biomedical Research Network in Cancer (CIBERONC), Av. Monforte de Lemos, 3-5. Pabellón 11. Planta 0, Madrid 28029, Spain
| | - Jelena Grahovac
- Experimental Oncology Department, Institute for Oncology and Radiology of Serbia, Belgrade, Serbia
| | - Didem Karakas
- Acibadem Mehmet Ali Aydinlar University, Department of Medical Biotechnology, Graduate School of Health Sciences, Istanbul, Turkey
| | - Giulia Lencioni
- Department of Biology, University of Pisa, Pisa, Italy; Fondazione Pisana per la Scienza, San Giuliano Terme, Pisa, Italy
| | - Sıla Sığırlı
- Acibadem Mehmet Ali Aydinlar University, Department of Medical Biotechnology, Graduate School of Health Sciences, Istanbul, Turkey
| | - Maarten F Bijlsma
- Amsterdam UMC Location University of Amsterdam, Laboratory of Experimental Oncology and Radiobiology, Amsterdam, the Netherlands; Cancer Center Amsterdam, Cancer Biology, Amsterdam, the Netherlands.
| |
Collapse
|
48
|
Wu J, Shao T, Tang Z, Liu G, Li Z, Shi Y, Kang Y, Zuo J, Zhao B, Hu G, Liu J, Ji W, Zhang L, Niu Y. Highly efficient construction of monkey blastoid capsules from aged somatic cells. Nat Commun 2025; 16:1130. [PMID: 39875393 PMCID: PMC11775175 DOI: 10.1038/s41467-025-56447-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2024] [Accepted: 01/16/2025] [Indexed: 01/30/2025] Open
Abstract
Blastoids-blastocyst-like structures created in vitro-emerge as a valuable model for early embryonic development research. Non-human primates stem cell-derived blastoids are an ethically viable alternative to human counterparts, yet the low formation efficiency of monkey blastoid cavities, typically below 30%, has limited their utility. Prior research has predominantly utilized embryonic stem cells. In this work, we demonstrate the efficient generation of blastoids from induced pluripotent stem cells and somatic cell nuclear transfer embryonic stem cells derived from aged monkeys, achieving an 80% formation efficiency. We also introduce a hydrogel-based microfluidics platform for the scalable and reproducible production of size-adjustable, biodegradable blastoid capsules, providing a stable 3D structure and mechanical protection. This advancement in the high-efficiency, scalable production of monkey blastoid capsules from reprogrammed aged somatic cells significantly enhances the study of embryonic development and holds promise for regenerative medicine.
Collapse
Affiliation(s)
- Junmo Wu
- State Key Laboratory of Primate Biomedical Research; Institute of Primate Translational Medicine, Kunming University of Science and Technology, Kunming, Yunnan, China
- Yunnan Key Laboratory of Primate Biomedical Research, Kunming, Yunnan, China
| | - Tianao Shao
- State Key Laboratory of Primate Biomedical Research; Institute of Primate Translational Medicine, Kunming University of Science and Technology, Kunming, Yunnan, China
- Yunnan Key Laboratory of Primate Biomedical Research, Kunming, Yunnan, China
| | - Zengli Tang
- State Key Laboratory of Primate Biomedical Research; Institute of Primate Translational Medicine, Kunming University of Science and Technology, Kunming, Yunnan, China
- Yunnan Key Laboratory of Primate Biomedical Research, Kunming, Yunnan, China
- Southwest United Graduate School, Kunming, Yunnan, China
| | - Gaojing Liu
- Key Laboratory of Genetic Evolution & Animal Models, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Zhuoyao Li
- State Key Laboratory of Primate Biomedical Research; Institute of Primate Translational Medicine, Kunming University of Science and Technology, Kunming, Yunnan, China
- Yunnan Key Laboratory of Primate Biomedical Research, Kunming, Yunnan, China
| | - Yuxi Shi
- State Key Laboratory of Primate Biomedical Research; Institute of Primate Translational Medicine, Kunming University of Science and Technology, Kunming, Yunnan, China
- Yunnan Key Laboratory of Primate Biomedical Research, Kunming, Yunnan, China
| | - Yu Kang
- State Key Laboratory of Primate Biomedical Research; Institute of Primate Translational Medicine, Kunming University of Science and Technology, Kunming, Yunnan, China
- Yunnan Key Laboratory of Primate Biomedical Research, Kunming, Yunnan, China
| | - Jiawei Zuo
- State Key Laboratory of Primate Biomedical Research; Institute of Primate Translational Medicine, Kunming University of Science and Technology, Kunming, Yunnan, China
- Yunnan Key Laboratory of Primate Biomedical Research, Kunming, Yunnan, China
- Southwest United Graduate School, Kunming, Yunnan, China
| | - Bo Zhao
- Key Laboratory of Genetic Evolution & Animal Models, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan, China
| | - Guangyu Hu
- State Key Laboratory of Primate Biomedical Research; Institute of Primate Translational Medicine, Kunming University of Science and Technology, Kunming, Yunnan, China
- Yunnan Key Laboratory of Primate Biomedical Research, Kunming, Yunnan, China
| | - Jiaqi Liu
- State Key Laboratory of Primate Biomedical Research; Institute of Primate Translational Medicine, Kunming University of Science and Technology, Kunming, Yunnan, China
- Yunnan Key Laboratory of Primate Biomedical Research, Kunming, Yunnan, China
| | - Weizhi Ji
- State Key Laboratory of Primate Biomedical Research; Institute of Primate Translational Medicine, Kunming University of Science and Technology, Kunming, Yunnan, China.
- Yunnan Key Laboratory of Primate Biomedical Research, Kunming, Yunnan, China.
| | - Lei Zhang
- State Key Laboratory of Primate Biomedical Research; Institute of Primate Translational Medicine, Kunming University of Science and Technology, Kunming, Yunnan, China.
- Yunnan Key Laboratory of Primate Biomedical Research, Kunming, Yunnan, China.
| | - Yuyu Niu
- State Key Laboratory of Primate Biomedical Research; Institute of Primate Translational Medicine, Kunming University of Science and Technology, Kunming, Yunnan, China.
- Yunnan Key Laboratory of Primate Biomedical Research, Kunming, Yunnan, China.
- Southwest United Graduate School, Kunming, Yunnan, China.
| |
Collapse
|
49
|
Huang Z, Zhou Y, Liu Y, Quan Y, Yin Q, Luo Y, Su Y, Zhou B, Zhang W, Zhu B, Ma Z. Advancing cellular transfer printing: achieving bioadhesion-free deposition via vibration microstreaming. LAB ON A CHIP 2025; 25:296-307. [PMID: 39655389 DOI: 10.1039/d4lc00601a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/29/2025]
Abstract
Cell transfer printing plays an essential role in biomedical research and clinical diagnostics. Traditional bioadhesion-based methods often necessitate complex surface modifications and offer limited control over the quantity of transferred cells. There is a critical need for a modification-free, non-labeling, and high-throughput cell transfer printing technique. In this study, an adhesion-free cellular transfer printing method based on vibration-induced microstreaming is introduced. By adjusting the volume of the microcavity, the number of cells transferred per microtiter well can be realized to the level of a single cell. Additionally, it allows for precise control of large-scale cellular spatial distribution, leading to the formation of biomimetic patterns. Moreover, the demonstrated biocompatibility and high throughput of this cell transfer printing method highlight its potential utility. The correspondence of the transferred cell amount to the vibration and frequencies allows the system to exhibit excellent tunability of the transferred cell amount and pattern. This bioadhesion-free cell transfer printing method holds promise for advancing cell manipulation in biomedical research and analysis.
Collapse
Affiliation(s)
- Ziyu Huang
- Joint Key Laboratory of the Ministry of Education, Institute of Applied Physics and Materials Engineering, University of Macau, Avenida da Universidade, Taipa, Macau 999078, China.
| | - Yinning Zhou
- Joint Key Laboratory of the Ministry of Education, Institute of Applied Physics and Materials Engineering, University of Macau, Avenida da Universidade, Taipa, Macau 999078, China.
| | - Yu Liu
- Joint Key Laboratory of the Ministry of Education, Institute of Applied Physics and Materials Engineering, University of Macau, Avenida da Universidade, Taipa, Macau 999078, China.
| | - Yue Quan
- Joint Key Laboratory of the Ministry of Education, Institute of Applied Physics and Materials Engineering, University of Macau, Avenida da Universidade, Taipa, Macau 999078, China.
| | - Qiu Yin
- Institute of Medical Robotics, School of Biomedical Engineering, Shanghai Jiao Tong University, No.800 Dongchuan Road, Shanghai 200240, China.
- State Key Laboratory of Mechanical System and Vibration, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Yucheng Luo
- Institute of Medical Robotics, School of Biomedical Engineering, Shanghai Jiao Tong University, No.800 Dongchuan Road, Shanghai 200240, China.
| | - Yimeng Su
- Institute of Medical Robotics, School of Biomedical Engineering, Shanghai Jiao Tong University, No.800 Dongchuan Road, Shanghai 200240, China.
| | - Bingpu Zhou
- Joint Key Laboratory of the Ministry of Education, Institute of Applied Physics and Materials Engineering, University of Macau, Avenida da Universidade, Taipa, Macau 999078, China.
| | - Wenming Zhang
- State Key Laboratory of Mechanical System and Vibration, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Benpeng Zhu
- School of Integrated Circuit, Wuhan National Laboratory for Optoelectronics, Huazhong University of Science and Technology, Wuhan 430074, China
| | - Zhichao Ma
- Institute of Medical Robotics, School of Biomedical Engineering, Shanghai Jiao Tong University, No.800 Dongchuan Road, Shanghai 200240, China.
| |
Collapse
|
50
|
Di Caprio N, Hughes AJ, Burdick JA. Programmed shape transformations in cell-laden granular composites. SCIENCE ADVANCES 2025; 11:eadq5011. [PMID: 39823334 PMCID: PMC11740954 DOI: 10.1126/sciadv.adq5011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/28/2024] [Accepted: 12/17/2024] [Indexed: 01/19/2025]
Abstract
Tissues form during development through mechanical compaction of their extracellular matrix (ECM) and shape morphing, processes that result in complex-shaped structures that contribute to tissue function. While observed in vivo, control over these processes in vitro to understand both tissue development and guide tissue formation has remained challenging. Here, we use combinations of mesenchymal stromal cell spheroids and hydrogel microparticles (microgels) with varied hydrolytic stability to fabricate programmable and dynamic granular composites that control compaction and tissue formation over time. Mixed microgel populations of varying stability provide a further handle to alter compaction, and the level of compaction guides the uniformity and level of ECM deposition within tissues. Last, spatially patterned granular composites of varying compaction enable shape transformations (i.e., bending/curvature) that are stable with culture and are predicted by finite element models.
Collapse
Affiliation(s)
- Nikolas Di Caprio
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA 19104, USA
- BioFrontiers Institute, University of Colorado Boulder, Boulder, CO 80303, USA
| | - Alex J. Hughes
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA 19104, USA
- Department of Cell & Developmental Biology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA USA 19104
| | - Jason A. Burdick
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA 19104, USA
- BioFrontiers Institute, University of Colorado Boulder, Boulder, CO 80303, USA
- Department of Chemical and Biological Engineering, University of Colorado Boulder, Boulder, CO 80303, USA
| |
Collapse
|